ArticlePDF Available

Hyperandrogenism and insulin resistance modulate gravid uterine and placental ferroptosis in PCOS-like rats

Authors:

Abstract and Figures

Previous studies in rats showed that maternal exposure to 5α-dihydrotestosterone (DHT) and insulin (INS) from gestational day 7.5 to 13.5 induces hyperandrogenism and insulin resistance (HAIR) and subsequently leads to placental insufficiency and fetal loss. We therefore hypothesized that maternal HAIR triggers ferroptosis in the uterus and placenta in association with fetal loss in pregnant rats. Compared with controls, we found that co-exposure to DHT and INS led to decreased levels of Gpx4 and glutathione (GSH), increased GSH+glutathione disulfide (GSSG) and malondialdehyde (MDA), aberrant expression of ferroptosis-associated genes (Acsl4, Tfrc, Slc7a11, and Gclc), increased iron deposition, and activated ERK/p38/JNK phosphorylation in the gravid uterus. However, in the placenta, DHT and INS exposure only partially altered the expression of ferroptosis-related markers (e.g., Gpx4, GSH+GSSG, MDA, Gls2 and Slc7a11 mRNAs, and phosphorylated p38 levels). In the uteri co-exposed to DHT and INS, we also observed shrunken mitochondria with electron-dense cristae, and increased Dpp4 mRNA expression. In contrast, in placentas co-exposed to DHT and INS we found decreased Dpp4 mRNA expression and increased Cisd1 mRNA expression. Further, DHT+INS-exposed pregnant rats exhibited decreased apoptosis in the uterus and increased necroptosis in the placenta. Our findings suggest that maternal HAIR causes the activation of ferroptosis in the gravid uterus and placenta, although this is mediated via different mechanisms operating at the molecular and cellular levels. Furthermore, our data suggest other cell death pathways may play a role in coordinating or compensating for HAIR-induced ferroptosis when the gravid uterus and placenta are dysfunctional.
Content may be subject to copyright.
https://doi.org/10.1530/JOE-20-0155
https://joe.bioscientifica.com © 2020 Society for Endocrinology
Printed in Great Britain
Published by Bioscientifica Ltd.
Journal of
Endocrinology
246:3 247–263
Y Zhang, M Hu etal. Uterine and placental
ferroptosis in PCOS
-20-0155
RESEARCH
Hyperandrogenism and insulin resistance
modulate gravid uterine and placental
ferroptosis in PCOS-like rats
YuehuiZhang1,*, MinHu2,3,*, WenyanJia1, GuoqiLiu1, JiaoZhang4, BingWang1, JuanLi2,3, PengCui2,5, XinLi2,6,7,
SusanneLager8, AmandaNancySferruzzi-Perri9, YanhuaHan1, SongjiangLiu1, XiaokeWu1, MatsBrännström10,
LinusRShao2 and HåkanBillig2
1Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Aliated Hospital, Heilongjiang University of
Chinese Medicine, Harbin, China
2Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg,
Gothenburg, Sweden
3Department of Traditional Chinese Medicine, The First Aliated Hospital of Guangzhou Medical University, Guangzhou, China
4Department of Acupuncture and Moxibustion, Second Aliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
5Department of Gynecology, Shuguang Hospital aliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
6Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
7Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
8Department of Women’s and Children’s Health, Uppsala University, Uppsala, Sweden
9Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
10Department of Obstetrics and Gynecology, Sahlgrenska University Hospital, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
Correspondence should be addressed to L R Shao: ruijin.shao@fysiologi.gu.se
*(Y Zhang and M Hu contributed equally to this work)
Abstract
Women with polycystic ovary syndrome (PCOS) have hyperandrogenism and insulin
resistance and a high risk of miscarriage during pregnancy. Similarly, in rats, maternal
exposure to 5α-dihydrotestosterone (DHT) and insulin from gestational day 7.5 to
13.5 leads to hyperandrogenism and insulin resistance and subsequently increased
fetal loss. A variety of hormonal and metabolic stimuli are able to trigger dierent
types of regulated cell death under physiological and pathological conditions. These
include ferroptosis, apoptosis and necroptosis. We hypothesized that, in rats, maternal
hyperandrogenism and insulin-resistance-induced fetal loss is mediated, at least in
part, by changes in the ferroptosis, apoptosis and necroptosis pathways in the gravid
uterus and placenta. Compared with controls, we found that co-exposure to DHT and
insulin led to decreased levels of glutathione peroxidase 4 (GPX4) and glutathione,
increased glutathione + glutathione disulde and malondialdehyde, aberrant expression
of ferroptosis-associated genes (Acsl4, Tfrc, Slc7a11, and Gclc), increased iron deposition
and activated ERK/p38/JNK phosphorylation in the gravid uterus. In addition, we
observed shrunken mitochondria with electron-dense cristae, which are key features of
ferroptosis-related mitochondrial morphology, as well as increased expression of Dpp4,
a mitochondria-encoded gene responsible for ferroptosis induction in the uteri of rats
co-exposed to DHT and insulin. However, in the placenta, DHT and insulin exposure only
partially altered the expression of ferroptosis-related markers (e.g. region-dependent
GPX4, glutathione + glutathione disulde, malondialdehyde, Gls2 and Slc7a11 mRNAs,
3
Key Words
fferroptosis
fmitochondria
fgravid uterus
fplacenta
fPCOS
246
Downloaded from Bioscientifica.com at 10/31/2021 03:48:54PM
via free access
https://doi.org/10.1530/JOE-20-0155
https://joe.bioscientifica.com © 2020 Society for Endocrinology
Published by Bioscientifica Ltd.
Printed in Great Britain
248
Uterine and placental
ferroptosis in PCOS
Y Zhang, M Hu etal. 246:3
Journal of
Endocrinology
and phosphorylated p38 levels). Moreover, we found decreased expression of Dpp4
mRNA and increased expression of Cisd1 mRNA in placentas of rats co-exposed to DHT
and insulin. Further, DHT + insulin-exposed pregnant rats exhibited decreased apoptosis
in the uterus and increased necroptosis in the placenta. Our ndings suggest that
maternal hyperandrogenism and insulin resistance causes the activation of ferroptosis
in the gravid uterus and placenta, although this is mediated via dierent mechanisms
operating at the molecular and cellular levels. Our data also suggest that apoptosis and
necroptosis may play a role in coordinating or compensating for hyperandrogenism
and insulin-resistance-induced ferroptosis when the gravid uterus and placenta are
dysfunctional.
Introduction
Polycystic ovary syndrome (PCOS) is a complex and
heterogeneous hormone-imbalance gynecological
disorder that is influenced by genetic, environmental,
and metabolic factors (Azziz et al. 2016). This disorder
affects approximately 4–21% of all adolescent and
reproductive-aged women and has a significant impact
on their reproduction (Liznevaetal. 2016). Women with
PCOS often suffer from hyperandrogenism/androgen
excess and insulin resistance (collectively termed; HAIR),
and they are at high risk for miscarriage and obstetric
complications during pregnancy (Bahri Khomami et al.
2019). Therapeutic interventions for different phenotypes
and disease-related pregnancy complications in women
with PCOS present a significant unmet medical need
(Rosenfield & Ehrmann 2016). Although it is thought that
maternal, placental and fetal defects all contribute to the
onset and progression of miscarriage in PCOS patients,
the pathogenesis of the pregnancy loss induced by HAIR
and its precise regulatory mechanisms are still significant
issues to be solved.
Ferroptosis is a recently described, iron-dependent
form of regulated necrosis induced by oxidative stress
and it is distinct from other established forms of cell
death, such as apoptosis and necroptosis (Choi et al.
2019), due to its unique morphological and biochemical
features (Dixon et al. 2012, Tang et al. 2019). Growing
evidence indicates that excessive or impaired ferroptosis
plays a causative role in a variety of pathological
conditions and diseases (Stockwell et al. 2017). It
appears that the outcome of ferroptosis is programmed
cell death, but which specific physiological processes or
pathological conditions and disorders lead to ferroptosis
activation remain poorly explored. The major molecular
mechanisms and signaling pathways that are involved in
the regulation of ferroptosis have been demonstrated in
in vivo and in vitro studies (Li et al. 2020). For example,
suppression of glutathione biosynthesis and subsequent
inhibition or degradation of glutathione peroxidase
4 (GPX4) activity, disturbed balance of iron homeostasis
and activation of the mitogen-activated protein kinase
(MAPK) signaling pathways all contribute to the initiation
and execution of ferroptosis (Xieetal. 2016). In addition
to ferroptosis, the alterations of apoptosis and necroptosis-
mediated signaling pathways have also been proposed as
the critical etiological factors of several human diseases
(Gudipatyetal. 2018). However, little is known about the
role of ferroptosis (Ng et al. 2019) in comparison with
other forms of programmed cell death such as apoptosis
(Spenceretal. 1996) in female reproduction.
Using rats, we have recently demonstrated that HAIR-
induced fetal loss is associated with uterine and placental
defects (Huetal. 2019b, Zhangetal. 2019b). In particular,
we exposed pregnant rats to 5α-dihydrotestosterone
(DHT) and insulin (INS) from gestational day (GD) 7.5
to 13.5 and found that this triggered many features of
PCOS (including HAIR) and lead to fetal loss. The fetal
loss was related to disrupted reactive oxygen species (ROS)
production in the uterus and placenta of rat dams with
induced HAIR. Maternal HAIR-induced fetal loss was
also associated with the inactivation of antioxidative
proteins in the gravid uterus and placenta, namely
nuclear factor erythroid 2-related factor 2 (Nrf2) and
superoxide dismutase 1 (Hu et al. 2019b, Zhang et al.
2019b), which play an inhibitory role in the ferroptosis
pathway (Xieetal. 2016, Tangetal. 2019). Moreover, the
mRNA expression of several other negative regulators of
ferroptosis such as heme oxygenase 1 (Ho1) (Tanget al.
2019) and metallothionein 1G (Mt1g) (Sun et al. 2016)
were downregulated in the gravid uterus after combined
maternal exposure to DHT and INS (Hu et al. 2019b).
Journal of Endocrinology
(2020) 246, 247–263
Downloaded from Bioscientifica.com at 10/31/2021 03:48:54PM
via free access
https://doi.org/10.1530/JOE-20-0155
https://joe.bioscientifica.com © 2020 Society for Endocrinology
Published by Bioscientifica Ltd.
Printed in Great Britain
249
Research
Y Zhang, M Hu etal. Uterine and placental
ferroptosis in PCOS
246:3
Journal of
Endocrinology
Increased circulating ROS levels have been observed in
both non-pregnant and pregnant rodents in which PCOS
features have been induced (Laietal. 2018, Zhang et al.
2019b). Elevated ROS production and decreased anti-
oxidative capacity has been observed in the ovarian
granulosa cells and leukocytes of PCOS patients
(Banulset al. 2017, Laiet al. 2018), and oxidative stress
is proposed to contribute to miscarriage and infertility
in women with PCOS (Agarwalet al. 2012, Schootsetal.
2018). It is, therefore, likely that the promotion of
pathologic oxidative stress and activation of ferroptosis
in the gravid uterus and placenta contribute to HAIR-
induced fetal loss in both animal models and humans.
Mitochondria play a protective role in the regulation
of glutathione-induced ferroptosis (Gao et al. 2019). In
women with PCOS and miscarriage, as well as in pregnant
PCOS-like rodents with fetal loss, there is mounting
evidence for mitochondrial abnormalities and oxidative
damage. For instance, decreased mitochondrial DNA copy
number is associated with the development and severity
of PCOS and several mitochondria-tRNA mutations are
seen in PCOS patients (Agarwalet al. 2012, Zhangetal.
2019a). In addition, aberrant expression of mitochondrial
biogenesis genes, oxidative phosphorylation and
anti-oxidative proteins are found in PCOS patients
who have recurrent miscarriage (Agarwal et al. 2012,
Zhang et al. 2019a), as well as in PCOS-like rodents
(Dingetal. 2019, Huetal. 2019a,b, Zhangetal. 2019b).
On the basis of these preclinical and clinical studies, we
hypothesized that maternal HAIR triggers impairments in
GPX4/glutathione-regulated lipid peroxidation and iron-
associated and mitochondria-mediated ferroptosis in the
gravid uterus and placenta resulting in increased fetal loss
during pregnancy.
The aim of this study was to determine whether
exposure to DHT and INS in pregnant rats (which
induces HAIR/PCOS (Huetal. 2019b, Zhangetal. 2019b))
leads to activation of the ferroptosis cascade, elevated
malondialdehyde (MDA, a marker of oxidative stress),
iron accumulation and perturbed mitochondrial function
in the uterus and placenta. Further, we conducted a
parallel analysis of the expression of genes and proteins
that are involved in necroptosis and apoptosis, two other
programmed cell death pathways that might contribute
to defects in the gravid uterus and the placenta. This
study is the first to report an association between HAIR
and different forms of regulated cell death in the gravid
uterus and placenta in vivo. Our findings indicate that
ferroptosis is one of the potential mechanisms by which
maternal HAIR leads to uterine and placental dysfunction
and at least partially explains the resultant fetal
loss observed.
Materials and methods
Ethics approval
All experiments were conducted in compliance with all
relevant local ethical regulations. Animal experiments
were approved and authorized by the Animal Care and
Use Committee of the Heilongjiang University of Chinese
Medicine, China (HUCM 2015-0112), and followed the
National Institutes of Health guidelines on the care and
use of laboratory animals.
Animals, experimental setting and tissue collection
Adult Sprague–Dawley female (n = 39) and male (n = 21)
rats were obtained from the Laboratory Animal Centre
of Harbin Medical University, Harbin, China. All animals
were health checked daily throughout the experiment and
were maintained in an environmentally controlled and
pathogen-free barrier facility on a standard 12 h light:12 h
darkness cycle at 22 ± 2 °C and 55–65% humidity and with
free access to normal diet and water. Before the experiment,
female rats (n = 9/group) were allowed to acclimatize for a
minimum of 7 days and then were monitored daily by
vaginal lavage to determine the stage of the estrous cycle
(Zhangetal. 2016). Pregnancy was achieved by housing
female rats on the night of proestrus with fertile males
of the same strain at a 2:1 ratio. Confirmation of mating
was defined by the presence of a vaginal plug and this
was considered as GD 0.5. Body weight of the rats was
recorded daily and rats were killed between 08:00 and
09:00 h on GD 14.5. All animal procedures in this study
were performed as described in our previous publications
(Huetal. 2019b, Zhangetal. 2019b).
To induce HAIR, pregnant rats were randomly
assigned to be intraperitoneally injected with DHT (1.66
mg/kg/day, suspended in sesame oil, Sigma-Aldrich)
and/or human recombinant INS (6.0 IU/day, diluted
in sterile saline, Eli Lilly Pharmaceuticals) or an equal
volume of saline and sesame oil as controls on GD 7.5
as previously described (Hu et al. 2019b, Zhang et al.
2019b). This therefore generated the following four study
groups: Control, DHT + INS, DHT, and INS. All animals
were treated for 7 consecutive days. The dose of DHT used
in our rats was chosen to mimic the hyperandrogenic
state in PCOS patients who have approximately
Downloaded from Bioscientifica.com at 10/31/2021 03:48:54PM
via free access
https://doi.org/10.1530/JOE-20-0155
https://joe.bioscientifica.com © 2020 Society for Endocrinology
Published by Bioscientifica Ltd.
Printed in Great Britain
250
Uterine and placental
ferroptosis in PCOS
Y Zhang, M Hu etal. 246:3
Journal of
Endocrinology
1.7-fold higher circulating DHT concentrations compared
to healthy controls (Fassnacht et al. 2003, Silfen et al.
2003). The dose of INS was chosen because it induces
metabolic disturbances including peripheral and uterine
insulin resistance in rats (Zhang et al. 2016, 2018). We
have previously shown that rats co-exposed to DHT and
INS during pregnancy had metabolic and endocrine
aberrations (HAIR) at GD 14.5 (Huetal. 2019b, Zhangetal.
2019b) that replicate the changes observed in pregnant
PCOS patients (Sir-Petermannetal. 2002, Maliqueoetal.
2013, Glintborg et al. 2018). The current investigation
used gravid uterine and placental tissues collected from
the same rats exposed to DHT and/or INS used in our
previous study (Zhangetal. 2019b), in which circulating
levels of androgens (testosterone, androstenedione,
dehydroepiandrosterone, and DHT), glucose tolerance
and fasting insulin, as well as fetal viability (litter size
and fetal loss per litter) were reported. Briefly these data
showed that rats co-exposed to DHT and INS had increased
androgen levels (testosterone, androstenedione, and
DHT) and worse insulin sensitivity, as well as decreased
litter size, with a corresponding increase in the percentage
of litters showing fetal loss. In addition, there was no
effect of DHT and/or insulin on maternal body weight
gain (Supplementary Fig. 1, see section on supplementary
materials given at the end of this article). On GD 14.5,
tissues, including the maternal uterus and placenta, as
well as fetuses were dissected. These were then either fixed
for morphological and immunohistochemical analyses
or immediately frozen in liquid nitrogen and stored at
70 °C for quantitative real-time PCR (qPCR) and Western
blot analyses. Only viable conceptuses (fetuses and
placentas) were analyzed further.
Detailed description of the methods including
the primers (Table 1) and qPCR analysis, Western blot
analysis, GPX4 immunostaining, Perls’ histochemical
reaction, transmission electron microscopy (TEM), and
quantification of glutathione, MDA and mitochondrial
open reading frame of the 12S rRNA-c (MOTS-c) used in
this study are provided in Supplementary files.
Data processing, statistical analysis, and graphical
representations
No statistical methods were used to pre-determine the
sample size. Data are presented as the means ± s.e.m., and
the sample size (n) is listed in the figure legends and
indicates the number of animals in each experiment.
Statistical analyses were performed using SPSS version
24.0 for Windows (SPSS Inc.). The normal distribution
of the data was tested with the Shapiro–Wilk test.
Differences between groups were analyzed by one-way
ANOVA followed by Tukey’s post-hoc test for normally
distributed data or the Kruskal–Wallis test for skewed data
(Supplementary Table 1). Body weight data were analyzed
by one-way ANOVA with repeated measures. Data were
not corrected for multiple testing. All P-values less than
0.05 were considered statistically significant.
Results
Because we were most interested in how HAIR induces
changes in ferroptosis as opposed to apoptosis and
necroptosis in gravid uterine and placental tissues, we have
mainly described the observations in DHT + INS-exposed
pregnant rats vs control pregnant rats subsequently.
Dierential regulation of GPX4 in the gravid uterus
and placenta exposed to DHT and INS
GPX4 is present in the cytoplasm, mitochondria and
nucleus of mammalian cells (Conrad et al. 2007).
Hence, we initially performed Western blot and
immunohistochemical analyses to characterize the
tissue and intracellular localization of GPX4 protein
in rat uterine and placental tissues. In the Western blot
analysis, the ~20-kDa band represents the cytosolic and
mitochondrial GPX4 protein in the rat testis, epididymis,
and ovary (Supplementary Fig. 2), as well as non-pregnant
and pregnant uteri (Supplementary Fig. 3A and Fig. 1A),
whereas the ~34-kDa band represents the nuclear GPX4
protein in the testis (Supplementary Fig. 2). Further
immunohistochemical studies showed that, while
positive immunostaining for cytosolic GPX4 was mainly
observed in luminal and glandular epithelial cells, GPX4
immunostaining was additionally localized to the nucleus
of stromal cells and myometrial smooth muscle cells in
non-pregnant rats (Supplementary Fig. 3B). In control
pregnant rats, GPX4 was localized to both the cytosol and
nucleus of different cells within the decidua, myometrium
and placenta (Fig. 1B1, B2, B3, and B4).
Although the significance of mitochondrial and
nuclear GPX4 remains to be determined (Forcina &
Dixon 2019), cytosolic GPX4 has been identified as a
central regulator of ferroptosis (Stockwelletal. 2017). We
thus evaluated GPX4 expression (Fig. 1A) and localization
(Fig. 1B, C, D, E, and F) in the gravid uterus and placenta
in rats exposed to DHT and INS. The Western blot
analysis revealed a significant decrease in uterine GPX4
Downloaded from Bioscientifica.com at 10/31/2021 03:48:54PM
via free access
https://doi.org/10.1530/JOE-20-0155
https://joe.bioscientifica.com © 2020 Society for Endocrinology
Published by Bioscientifica Ltd.
Printed in Great Britain
251
Research
Y Zhang, M Hu etal. Uterine and placental
ferroptosis in PCOS
246:3
Journal of
Endocrinology
abundance in DHT + INS-exposed pregnant rats (Fig.
1A). Consistent with this, there appeared to be weaker
immunoreactivity of GPX4 in the cytosolic compartments
of decidualized stromal and smooth muscle cells in the
uterus of DHT + INS-exposed pregnant rats (Fig. 1C1
and C2). Although there was no significant difference
in uterine GPX4 abundance by Western blot analysis in
pregnant rats treated alone with DHT or INS (Fig. 1A),
the number of cytosolic and/or nuclear GPX4-positive
uterine cells was decreased when compared to controls
using immunostaining (Fig. 1D1, D2 and E2). Similarly,
while GPX4 protein abundance was unchanged in the
placenta of DHT + INS-exposed pregnant rats by Western
blot analysis (Fig. 1A), cytosolic GPX4 immunoreactivity
appeared to be lower in the junctional and labyrinth zones
in DHT + INS-exposed pregnant rats when examined by
immunohistochemistry (Fig. 1C3 and C4). In particular,
GPX4 immunostaining was no longer localized to the
nuclei of spongiotrophoblast, glycogen, cytotrophoblast
and syncytiotrophoblast cells of DHT + INS-exposed
Table 1 Primer sequences used for qPCR measurement.
Gene Primer sequence (5-3)Reference sequence Product size (bp)
Slc1a5 Forward TCGGGACCTCTTCTAGCTCT NM_175758.3 90
Reverse TGAACCGGCTGATGTGTTTG
Acsl4 Forward CTCCTGCTTTACCTACGGCT NM_053623.1 97
Reverse ACAATCACCCTTGCTTCCCT
Gls2 Forward GGCCAAGTCAAACCCAGATC NM_001270786.1 153
Reverse TAGTCGGTGCCTAAGGTGC
Cs Forward AGTGCCAGAAACTGCTACCT NM_130755.1 117
Reverse GTGAGAGCCAAGAGACCTGT
Gclc Forward AAGCCATAAACAAGCACCCC NM_012815.2 116
Reverse CGGAGATGGTGTGTTCTTGTC
Gss Forward ATGCCGTGGTGCTACTGATT NM_012962.1 107
Reverse TCTTCGGCGGATTACATGGA
Tfrc Forward AGGCTCCTGAGGGTTATGTG NM_022712.1 204
Reverse AGATGAGGACACCAATTGCA
Ireb2 Forward TGTTTGAAGAAGCCGACCTG NM_022863.2 97
Reverse ACTCCCCACCCAAGAATTCC
Slc7a11 Forward GTGCCCGGATCCAGATTTTC NM_001107673.2 270
Reverse TGATGGCCATAGAGATGCAGA
Cisd1 Forward GCTAAAGAGAGTCGCACCAAAG NM_001106385.2 113
Reverse CGGCAATACACGGCCTTATC
Dpp4 Forward GGCTGGTGCGGAAGATTTA NM_012789.1 135
Reverse GACCTGTTCGGGTTTCCTATC
Bcl2 Forward TTGCAGAGATGTCCAGTCAG NM_016993.1 125
Reverse GAACTCAAAGAAGGCCACAATC
Bcl-xl Forward GGTGGTTGACTTTCTCTCCTAC NM_031535.2 116
Reverse TCTCCCTTTCTGGTTCAGTTTC
Bax Forward GATGGCCTCCTTTCCTACTTC NM_017059.2 96
Reverse CTTCTTCCAGATGGTGAGTGAG
Bak Forward GATCGCCTCCAGCCTATTTAAG NM_053812.1 115
Reverse CAGGAAGCCAGTCAAACCA
Casp3 Forward GACTGGAAAGCCGAAACTCT NM_012922.2 97
Reverse TGCCATATCATCGTCAGTTCC
Mlkl Forward GGAACTGCTGGATAGAGACAAG XM_008772570.2 117
Reverse CTGATGTTTCCGTGGAGTGT
Ripk1 Forward CAGGTACAGGAGTTTGGTATGG NM_001107350.1 108
Reverse TGTATGGCATGGTGGGTATG
Ripk3 Forward ACTGAGAGGAGAGGAAAGGAAG NM_139342.1 107
Reverse CTGGAGGGTAGAGTATGTGGAA
Gapdh Forward TCTCTGCTCCTCCCTGTTCTA NM_017008.4 121
Reverse GGTAACCAGGCGTCCGATAC
Acsl4, acyl-CoA synthetase long-chain family member 4; Bak, bcl-2 homologous antagonist killer; Bax, bcl-2-like protein 4; Bcl2, b-cell lymphoma 2; Bcl-xl,
b-cell lymphoma-extra large; Casp3, caspase 3; Cisd1, CDGSH iron sulfur domain 1; Cs, citrate synthase; Dpp4, dipeptidylpeptidase 4; Gapdh,
glyceraldehyde-3-phosphate dehydrogenase; Gclc, glutamate-cysteine ligase catalytic subunit; Gls2, glutaminase 2; Gss, glutathione synthetase; Ireb2, iron
responsive element binding protein 2; Mlkl, mixed lineage kinase domain like pseudokinase; Ripk1, receptor interacting serine/threonine kinase 1; Slc1a5,
solute carrier family 1 member 5; Slc7a11, solute carrier family 7 member 11; Tfrc, transferrin receptor.
Downloaded from Bioscientifica.com at 10/31/2021 03:48:54PM
via free access
https://doi.org/10.1530/JOE-20-0155
https://joe.bioscientifica.com © 2020 Society for Endocrinology
Published by Bioscientifica Ltd.
Printed in Great Britain
252
Uterine and placental
ferroptosis in PCOS
Y Zhang, M Hu etal. 246:3
Journal of
Endocrinology
pregnant rats compared to controls (Fig. 1C3 and
C4). While cytosolic GPX4 immunoreactivity was
decreased in both spongiotrophoblasts and glycogen
cells, nuclear GPX4 immunoreactivity was absent in
spongiotrophoblasts cells in pregnant rats treated with
DHT alone (Fig. 1D3) and INS alone (Fig. 1E3). Similar
to that of controls (Fig. 1B4), GPX4 immunoreactivity
was found in the placental labyrinth zone in pregnant
rat placentas exposed to DHT or INS alone (Fig. 1D4 and
F4). No obvious GPX4 immunostaining was evident in
the uterine and placental tissue sections using the same
concentration of isotype-matched rabbit IgG instead of
the primary GPX4 antibody (Fig. 1F1, F2, F3, and F4).
Dierential regulation of glutathione content in the
gravid uterus and placenta exposed to DHT and INS
GPX4 uses glutathione as a substrate in its peroxidase
reaction cycle (Conradetal. 2007) and glutathione depletion
is one of the key triggers for ferroptosis (Xieet al. 2016,
Stockwelletal. 2017). Therefore, we measured the levels of
glutathione and glutathione + glutathione disulfide in the
gravid uterus and placenta in rats exposed to DHT and INS.
As shown in Fig. 2A, co-exposure of rats to DHT and INS
decreased glutathione levels in the gravid uterus, but not
in the placenta, while increased glutathione + glutathione
disulfide levels were detected in both tissues.
Figure1
Regulation and localization of GPX4 protein in pregnant rats exposed to DHT and/or INS at GD 14.5. Western blot analysis of GPX4 protein expression in
the uterus and placenta (A, n= 9/group). In all plots, values are expressed as means ± s.e.m. Statistical P values for selected comparisons are indicated as
**P < 0.01. Histological analysis by GPX4 immunostaining in the gravid uterus (Mt and Md) and placenta (Jz and Lz) (B1–F4). A negative control was
performed by using the same concentration of isotype-matched rabbit IgG instead of the primary antibody. Only minimal cytoplasmic background
staining was observed (F, and F1–4). Tissue sections were counterstained with methyl green. Images are representative of 810 tissue replicates per
group. Mt, mesometrial triangle; SA, spiral artery; Md, mesometrial decidua; Jz, junctional zone (maternal side); Gc, glycogen cells; Sp, spongiotrophoblast
cells; Lz, labyrinth zone (fetal side); Cy, cytotrophoblast; Sy, syncytiotrophoblast; Mv, maternal blood vessel; Fv, fetal blood vessel. Small (100 μm) and big
(50 μm) scale bars are indicated in the photomicrographs. DHT, 5α-dihydrotestosterone; INS, insulin.
Downloaded from Bioscientifica.com at 10/31/2021 03:48:54PM
via free access
https://doi.org/10.1530/JOE-20-0155
https://joe.bioscientifica.com © 2020 Society for Endocrinology
Published by Bioscientifica Ltd.
Printed in Great Britain
253
Research
Y Zhang, M Hu etal. Uterine and placental
ferroptosis in PCOS
246:3
Journal of
Endocrinology
Alterations in glutathione and glutathione + glutathione
disulfide levels were found in the gravid uterus in pregnant
rats exposed to INS alone and levels of glutathione were
lower in both the uterus and placenta in pregnant rats
treated with DHT alone (Fig. 2A).
Alterations in ferroptosis-related gene expression in
the gravid uterus and placenta with DHT and INS
Next, we examined whether maternal exposure to DHT
and INS alters the expression of pro-ferroptosis (Slc1a5,
Acsl4, Gls2, Cs, Tfrc and Ireb2) or anti-ferroptosis (Slc7a11,
Gclc, and Gss) genes (Dai et al. 2020) in the uterus and
placenta. In pregnant DHT + INS-exposed rats, uterine
Acsl4, Slc7a11 and Gclc mRNAs were decreased, while Tfrc
mRNA was increased (Fig. 2B). In comparison with the
control uterus, maternal exposure to DHT alone decreased
Cs, Ireb2, Slc7a11, Gclc and Gss mRNA expression, whereas
exposure to INS alone increased Gls2 and Tfrc mRNAs
and decreased Slc7a11 mRNA expression (Fig. 2B). qPCR
analysis also showed that Gls2 mRNA expression was
increased and Slc7a11 mRNA expression was decreased
in the placenta after maternal co-exposure to DHT and
INS. In comparison with the control placenta, exposure
to DHT alone decreased Cs, Slc7a11 and Gss mRNA
expression, whereas exposure to INS alone decreased Gclc
and Gss mRNAs in parallel to increased Tfrc and Slc7a11
mRNA expression (Fig. 2B).
Alterations in MDA levels in the gravid uterus and
placenta with DHT and INS
Given that one of the key consequences of ferroptosis
is elevated lipid peroxidation (Dixon et al. 2012,
Figure2
Alteration of glutathione,
glutathione + glutathione disulde, ferroptosis-
related gene expression, and MDA in pregnant
rats exposed to DHT and/or INS at GD 14.5. ELISA
analysis of glutathione (the reduced state),
glutathione + glutathione disulde, and MDA in
the uterus and placenta (A, n= 8/group). qPCR
analysis of uterine and placental genes involved in
modulating ferroptosis (B, n= 78/group). In all
plots, values are expressed as means ± s.e.m.
Statistical P values for selected comparisons
are indicated as *P < 0.05, **P < 0.01, and
***P < 0.001. DHT, 5α-dihydrotestosterone;
INS, insulin.
Downloaded from Bioscientifica.com at 10/31/2021 03:48:54PM
via free access
https://doi.org/10.1530/JOE-20-0155
https://joe.bioscientifica.com © 2020 Society for Endocrinology
Published by Bioscientifica Ltd.
Printed in Great Britain
254
Uterine and placental
ferroptosis in PCOS
Y Zhang, M Hu etal. 246:3
Journal of
Endocrinology
Tangetal. 2019), we next examined the impact of DHT and
INS on the levels of MDA, a marker of lipid peroxidation
(Gaweletal. 2004), in the gravid uterus and placenta. As
shown in Fig. 2C, maternal co-exposure to DHT and INS
resulted in increased MDA levels in both the gravid uterus
and placenta. However, there were no significant changes
in MDA levels between the DHT-exposed rats or the
INS-exposed rats and control rats.
Alterations in intracellular iron deposition in the
gravid uterus and placenta with DHT and INS
Because disturbed iron transport and impaired metabolism
within cells/tissues results in ferroptosis (Galaris etal. 2019),
whether chronic exposure to DHT and INS can modulate
tissue iron deposition was also examined. Perls’
histochemical reaction showed specific cytoplasmic
and granular iron storage in rat uterine epithelial and
decidualized stromal cells on GD 6, which is prior to the
induction of HAIR (Supplementary Fig. 3A1 and A2).
As compared to control pregnant rats (Fig. 3A1, A2, A3
and Supplementary Fig. 4B1, B2), iron accumulation was
increased in the external muscle layer, the mesometrial
triangle, as well as in the decidua of DHT + INS-exposed
pregnant rats (Fig. 3B1, B2, B3 and Supplementary Fig.
4B3, B4). Similarly, a significant increase in iron storage
in the mesometrial triangle was also observed in DHT-
exposed rat dams (Fig. 3C1 and C2). In the mesometrial
Figure3
Iron deposition in the uterus and placenta of
pregnant rats exposed to DHT and/or INS at GD
14.5. Gravid uterine and placental tissues from
pregnant rats treated with vehicle (A1–5),
DHT + INS (B1–5), DHT (C1–5), or INS (D1–5) are
shown. The sections were stained by DAB-
enhanced Perls’ staining for iron accumulation.
Yellow arrowheads indicate iron-positive staining.
Images are representative of eight tissue
replicates per group. Mt, mesometrial triangle;
Md, mesometrial decidua; Jz, junctional zone
(maternal side); Lz, labyrinth zone (fetal side).
Scale bars (100 μm) are indicated in the
photomicrographs. DHT, 5α-dihydrotestosterone;
INS, insulin.
Downloaded from Bioscientifica.com at 10/31/2021 03:48:54PM
via free access
https://doi.org/10.1530/JOE-20-0155
https://joe.bioscientifica.com © 2020 Society for Endocrinology
Published by Bioscientifica Ltd.
Printed in Great Britain
255
Research
Y Zhang, M Hu etal. Uterine and placental
ferroptosis in PCOS
246:3
Journal of
Endocrinology
decidua, granular and cytoplasmic iron-positive staining
was absent in the DHT-exposed rats (Fig. 3C3) but was
barely detectable in the INS-exposed rats (Fig. 3C3 and
D3). However, no iron-positive staining was found in
the placental junctional zone in any of the experimental
groups (Fig. 3A4, B4, C4, and D4), while intense iron-
positive staining was consistently detected in immature
erythrocytes within the placental labyrinth zone in all
experimental groups (Fig. 3A5, B5, C5 and D5). These
results indicate that the amount of deposited iron was
elevated, especially in the gravid uterus, following
exposure to DHT and/or INS.
Alterations in the MAPK signaling pathway in the
gravid uterus and placenta with DHT and INS
Taking into consideration that the MAPK signaling
pathway, including ERK, p38, and c-JUN NH2-terminal
kinase (JNK), is involved in the execution of ferroptosis
in other cells (Xie et al. 2016), we evaluated whether
co-exposure to DHT and INS may be linked to activation
of the MAPK signaling pathway in the gravid uterus
and placenta. As shown in Fig. 4A, in the gravid uterus,
maternal DHT + INS exposure resulted in an increased
abundance of phosphorylated ERK1/2 (p-ERK1/2) and
decreased total ERK1/2, which subsequently resulted in an
increased p-ERK1/2:ERK1/2 ratio. Moreover, both p-JNK
and total JNK protein abundance were increased, whereas
the p-JNK:JNK ratio remained unchanged in the gravid
uterus of DHT + INS-exposed rats (Fig. 4A). Additionally,
a similar increase in p-p38 protein abundance and the
p-p38:p38 ratio was observed in both the gravid uterus
(Fig. 4A) and placenta (Fig. 4B) after maternal co-exposure
to DHT and INS. These results indicate that both ERK1/2
and JNK signaling are only activated in the gravid
uterus, whereas p38 signaling is activated in both the
gravid uterus and placenta after maternal co-exposure to
DHT and INS.
Figure4
Changes in the expression of proteins involved in
the ferroptosis-related MAPK signaling pathway in
pregnant rats exposed to DHT and/or INS at GD
14.5. Western blot analysis of ERK, p38, and JNK
protein expression and their phosphorylated
forms in the uterus and placenta (n= 9/group). In
all plots, values are expressed as means ± s.e.m.
Statistical P values for selected comparisons
are indicated as *P < 0.05, **P < 0.01, and
***P < 0.001. DHT, 5α-dihydrotestosterone;
INS, insulin.
Downloaded from Bioscientifica.com at 10/31/2021 03:48:54PM
via free access
https://doi.org/10.1530/JOE-20-0155
https://joe.bioscientifica.com © 2020 Society for Endocrinology
Published by Bioscientifica Ltd.
Printed in Great Britain
256
Uterine and placental
ferroptosis in PCOS
Y Zhang, M Hu etal. 246:3
Journal of
Endocrinology
Changes in mitochondrial morphology are associated
with changes in mitochondria-encoded gene and
protein expression in the gravid uterus and placenta
with DHT and INS
By TEM (Fig. 5 and Supplementary Fig. 5), we found
shrunken mitochondria with numerous electron-dense
cristae or absent cristae in the gravid uterus of DHT + INS-
exposed rats (Fig. 5B1 arrows) compared to controls (Fig.
5A1). Further, mitochondria were swollen and collapsed
with poorly defined tubular cristae in the gravid uterus
of rat exposed to DHT and/or INS (Fig. 5B1, C1, and
D1). Our TEM findings of the uterus in DHT + INS rats
are consistent with ferroptosis-related mitochondrial
morphology (Dixonet al. 2012, Xieet al. 2016, Liet al.
2020). Treatment with DHT or INS also reduced the
number of mitochondrial cristae in the uterus (Fig. 5C1
and D1). Ultrastructural analysis of the placenta showed
that mitochondria in the trophoblast of the junctional
zone were significantly affected by maternal exposure to
DHT and/or INS (Fig. 5A2, B2, C2, and D2). For instance,
mitochondria showed blebbing, few or no tubular
cristae and decreased electron density in all treatment
groups (Fig. 5B2, C2, and D2). However, there was little
mitochondrial damage observed in the trophoblast of the
placental labyrinth zone in all treatment groups compared
to controls (Fig. 5A3, B3, C3, and D3).
Based on these morphological observations, the
expression of known mitochondria-encoded genes
(Cisd1, an anti-ferroptosis gene and Dpp4, a pro-
ferroptosis gene (Stockwell et al. 2017, Tang et al.
2019)) and protein (MOTS-c, an enhancer of insulin
sensitivity (Kimetal. 2017)) were analyzed by qPCR and
ELISA. In the pregnant rat uterus, DHT + INS-exposure
decreased Cisd1 mRNA expression, increased Dpp4
mRNA expression and decreased the MOTS-c protein
level (Fig. 5E and F upper panel). In contrast, we found
significantly higher uterine Cisd1 and Dpp4 mRNA
expression in INS-exposed pregnant rats (Fig. 5E upper
panel), but unchanged uterine MOTS-c protein levels in
DHT-exposed pregnant rats compared to controls (Fig.
5F upper panel). In the placenta, Cisd1 mRNA expression
was increased and Dpp4 mRNA expression was decreased
in DHT + INS-exposed pregnant rats compared to
controls (Fig. 5E lower panel). A decrease in placental
Dpp4 mRNA expression was also observed in INS-
exposed pregnant rats (Fig. 5E lower panel). However,
there was no significant difference in MOTS-c protein
levels in the placenta between any of the experimental
groups (Fig. 5F lower panel).
Aberrant regulation of necroptosis-related and
anti-/pro-apoptosis-related gene and protein
expression in the gravid uterus and placenta with
DHT and INS
Different types of cell death are seen in uterine and
placental tissue during healthy and pathological
pregnancy (Welsh 1993, Sharp et al. 2010). To extend
our observations on the effect of maternal DHT and INS
treatment on ferroptosis and mitochondrial impairment,
we analyzed the expression of necroptosis (Mlkl, Ripk1
and Ripk3), anti-apoptosis (Bcl2 and Bcl-xl) and pro-
apoptosis (Bax, Bak, Casp3 and cleaved caspase-3) mRNAs
and proteins (Xieetal. 2016, Choietal. 2019, Tangetal.
2019) in the gravid uterus and placenta. As shown in Fig.
6A, DHT + INS-exposure significantly decreased uterine
Ripk1 mRNA expression, while uterine Mlkl and Ripk3
mRNAs were increased by DHT and/or INS exposure when
compared to control pregnant rats (Fig. 6A upper panel).
Furthermore, co-exposure to DHT and INS increased
Bcl-xl and Bax mRNA expression in the gravid uterus,
with similar increases in these genes seen in DHT-exposed
and/or INS-exposed pregnant rats compared to controls
(Fig. 6B upper panel). Gravid uterine Bcl2 mRNA expression
was not altered by co-exposure to DHT and INS; however,
it was increased by DHT and decreased by INS when
compared to control pregnant rats. In DHT + INS-exposed
pregnant rats, Casp3 mRNA expression and cleaved
caspase-3 protein abundance were decreased in the gravid
uterus (Fig. 6B upper panel and C). In contrast, in the
placenta we found that both Ripk1 and Ripk3 mRNAs were
increased in DHT + INS-exposed pregnant rats compared
to controls (Fig. 6A lower panel). Furthermore, maternal
co-exposure to DHT and INS increased placental Bcl-xl, Bax
and Bak mRNA expression (Fig. 6B lower panel). Of note,
placental Bcl2 mRNA expression was also increased in
DHT + INS-exposed rats and most significantly increased in
the INS alone exposure. However there was no significant
effect of the DHT alone exposure on placental Bcl2 mRNA
level. There were, however, no changes in Casp3 mRNA
expression or cleaved caspase-3 protein abundance in
the placenta (Fig. 6B lower panel and C). Lastly, similar
increases in placental Bcl-xl, Bax, Bak and Casp3 mRNAs
were seen in DHT-exposed and/or INS-exposed pregnant
rats compared to controls (Fig. 6B lower panel).
Discussion
Because PCOS patients frequently suffer from miscarriage
and infertility (Bahri Khomamietal. 2019), it is important
Downloaded from Bioscientifica.com at 10/31/2021 03:48:54PM
via free access
https://doi.org/10.1530/JOE-20-0155
https://joe.bioscientifica.com © 2020 Society for Endocrinology
Published by Bioscientifica Ltd.
Printed in Great Britain
257
Research
Y Zhang, M Hu etal. Uterine and placental
ferroptosis in PCOS
246:3
Journal of
Endocrinology
Figure5
Electron microscopy and mitochondria-mediated ferroptosis-related gene and protein expression in pregnant rats exposed to DHT and/or INS at GD
14.5. Mitochondrial ultrastructural defects in the uterus (A1, B1, C1, and D1, mesometrial decidua) and placenta (junctional (A2, B2, C2, and D2) and
labyrinth zones (A3, B3, C3, and D3)). Images are representative of two tissue replicates. Md, mesometrial decidua; Jz, junctional zone (maternal side); Lz,
labyrinth zone (fetal side). Red asterisks indicate mitochondria, and white arrows indicate shrunken mitochondria with electron-dense cristae. Scale bars
(500 nm) are indicated in the photomicrographs. qPCR analysis of mitochondrial genes involved in modulating ferroptosis (E, n= 8/group). ELISA analysis
of MOTS-c content (F, n= 8/group). In all plots, values are expressed as means ± s.e.m. Statistical P values for selected comparisons are indicated as
*P < 0.05, **P < 0.01, and ***P < 0.001. DHT, 5α-dihydrotestosterone; INS, insulin.
Downloaded from Bioscientifica.com at 10/31/2021 03:48:54PM
via free access
https://doi.org/10.1530/JOE-20-0155
https://joe.bioscientifica.com © 2020 Society for Endocrinology
Published by Bioscientifica Ltd.
Printed in Great Britain
258
Uterine and placental
ferroptosis in PCOS
Y Zhang, M Hu etal. 246:3
Journal of
Endocrinology
to understand the molecular mechanisms through which
HAIR affects tissues such as the gravid uterus and placenta.
Until now, there have been no reports exploring the
relationship between PCOS and regulated cell death in
the uterus and placenta. Our results thus fill an important
clinically relevant knowledge gap by experimentally
demonstrating that maternal HAIR can cause the
activation of ferroptosis in the gravid uterus and placenta,
although this is mediated through different molecular
and cellular mechanisms. We propose that alterations in
the ferroptosis pathway in the uterus and placenta due to
maternal HAIR likely contribute to impaired fetal survival
seen in experimental animal models and future work is
required to assess whether this is also the case in women
with PCOS.
In mammals, GPX4 plays a major role in antioxidant
defense by regulating responses to oxidative stress.
Furthermore, loss of function of GPX4 protein and
depletion of GSH levels are the key mechanisms for
triggering ferroptosis (Dixon et al. 2012, Stockwell et al.
2017). In vivo knockout studies have shown that mice
lacking the entire GPX4 gene experience early embryonic
lethality (Imai et al. 2003) and that GPX4-deficient
male mice are infertile (Schneideretal. 2009). Although
the presence of GPX4 has been shown in uteri from
cows (Ramos et al. 2015, Baithalu et al. 2017) and pigs
(Dalto et al. 2015), the localization and physiological
role of GPX4 has not been demonstrated in human and
rodent reproductive tissues, including the uterus. Here,
we show that GPX4 is widely expressed in the non-
pregnant and pregnant rat uteri, including decidualized
stromal cells. The data presented showing the differential
cellular GPX4 localization in the uterus is consistent with
variations in the compartmentalization of GPX4 between
the cytosol and nucleus in different cells (Conrad et al.
2007). Furthermore, GPX4 is down-regulated in the
gravid uterus by maternal exposure to DHT and INS.
Correspondingly, the levels of glutathione are decreased
and glutathione+glutathione disulfide levels are increased
in the uterus by DHT and INS co-exposure. Taken together,
these results suggest that maternal HAIR disrupts the
GPX4-glutathione regulatory axis and can result in the
induction of ferroptosis in the uterus during pregnancy.
The finding that glutathione levels in the uterus were
lowest in the INS-only treated rats which also showed a
non-significant reduction in GPX4 suggests that other
signaling pathways and factors such as the transcription
factors Nrf1 and Nrf2 (Lu 2009) might be altered by the
treatments and contribute to the resultant changes in
glutathione status and should be investigated for causality
in the future (also in the placenta of DHT and/or INS-
exposed dams). Indeed, we have previously found altered
abundance of antioxidants in the uterus of pregnant rats
exposed to DHT and/or INS (Huetal. 2019b, Zhangetal.
2019b). Consistent with previous work on the human
placenta (Mistry et al. 2008, 2010), the present study
Figure6
The regulatory pattern of necroptosis-related and pro-/anti-apoptosis-related gene and protein expression in pregnant rats exposed to DHT and/or INS
at GD 14.5. qPCR analysis of Mlkl, Ripk1, Ripk3, Bcl2, Bcl-xl, Bax, Bak, and Casp3 mRNA in the uterus and placenta (A and B, n= 8/group). Western blot
analysis of cleaved caspase-3 protein expression in the uterus and placenta (C, n= 9/group). In all plots, values are expressed as means ± s.e.m. Statistical
P values for selected comparisons are indicated as *P < 0.05, **P < 0.01, and ***P < 0.001. DHT, 5α-dihydrotestosterone; INS, insulin.
Downloaded from Bioscientifica.com at 10/31/2021 03:48:54PM
via free access
https://doi.org/10.1530/JOE-20-0155
https://joe.bioscientifica.com © 2020 Society for Endocrinology
Published by Bioscientifica Ltd.
Printed in Great Britain
259
Research
Y Zhang, M Hu etal. Uterine and placental
ferroptosis in PCOS
246:3
Journal of
Endocrinology
shows that the GPX4 protein is highly expressed in the rat
placenta during pregnancy. Although analysis of whole
placental homogenates showed no significant change
in GPX4 levels, immunolocalization revealed a loss of
GPX4 in specific cell types in the placenta (the glycogen
and spongiotrophoblast cells) in response to maternal
co-exposure to DHT and INS. The more minor alterations
in GPX4 abundance, combined with the high levels of
glutathione + glutathione disulfide and absence of changes
in glutathione levels in the placenta, suggest that maternal
HAIR induces ferroptosis to a lesser extent in the placenta
compared to the gravid uterus. GPX4 is known to protect
cells/tissues against lipid peroxidation by inhibiting
lipid-associated hydroperoxides (Conrad et al. 2007).
In addition, genetically ablating or inducing decreased
GPX4 expression leads to the activation of ferroptosis
(Friedmann Angelietal. 2014, Chenetal. 2015). Together,
our data therefore suggest that HAIR-induced ferroptosis is
mediated by both dysregulation of GPX4 expression and
aberrant increases in lipid peroxidation. The induction of
uterine and placental ferroptosis by maternal exposure to
DHT and INS may be a novel mechanism contributing
to the malfunction of those tissues and hence impaired
fetal development during pregnancy. However, how
maternal HAIR-mediated uterine and placental ferroptosis
compromises the growth and development of the fetus
is not clear at this time and should be the subject of
future investigations. Moreover, future work should be
employed to assess whether the activation of ferroptosis,
lipid peroxidation and poor fetal outcomes by maternal
HAIR may be preventable by antioxidant administration.
Iron can serve as an essential signaling molecule
that modulates diverse physiological processes and
iron homeostasis is required for the normal growth
and development of the placenta and fetus during
pregnancy (Cao & Fleming 2016, Ng et al. 2019). By
Perls’ histochemical reaction, we found a considerable
proportion of uterine epithelial and decidualized stromal
cells stained positively for iron storage on GD 6. These
data are consistent with a previous report showing the
cellular expression of ferritin heavy chain, a component
of the multi-subunit iron-binding protein ferritin, in
the uterus during early pregnancy (Zhu et al. 1995). An
extensive body of evidence indicates that, while iron
deficiency is linked to abnormal pregnancy (Ng et al.
2019) and increased risk of fetal death (Guoet al. 2019),
iron overload is associated with the manifestation of
PCOS (Escobar-Morreale 2012). Previous findings by
Kim and colleagues indicate that increased circulating
iron levels are associated with metabolic abnormalities,
including HAIR in PCOS patients (Kim et al. 2014).
Several studies have demonstrated that, in addition to
its antioxidative property, Ho1 is a critical regulator for
mobilization of intracellular pools of free iron (Poss &
Tonegawa 1997, Kovtunovychetal. 2010). More recently,
we have demonstrated that maternal co-exposure to
DHT and INS suppresses Ho1 mRNA expression in the
gravid uterus, but not in the placenta (Hu et al. 2019b,
Zhangetal. 2019b). While the uptake of transferrin-bound
iron, a major maternal iron source for placental transfer,
is mainly mediated through iron import proteins such as
transferrin receptor 1 (TFR1, TFRc) (Cao & Fleming 2016),
a specific ferroptosis marker (Fengetal. 2020), our results
show that combined exposure to DHT and INS increases
Tfrc mRNA expression in association with increased iron
deposition in the gravid uterus. Further, we have provided
ultrastructural evidence that shrunken mitochondria
with numerous electron-dense cristae, a key feature of
ferroptosis-related mitochondrial morphology, are present
in the gravid uterus. However, the placentas of the same
animals exhibited increased mRNA expression of Cisd1, a
mitochondrial iron export factor, and no change in Tfrc
mRNA or iron accumulation. Ferroptosis can be induced
by excessive accumulation of free iron in tissues and cells
(Galariset al. 2019) and our findings support the notion
that, in response to exposure to DHT and INS, aberrant
iron accumulation and activation of ferroptosis occurs
in the gravid uterus but not in the placenta. Given the
fact that whether or not mitochondria are involved in
ferroptosis is still under debate (Gaoet al. 2019), further
investigations are needed to determine which cellular
compartments contribute to the defective utilization
of iron and increased ferroptosis observed in the gravid
uterus under conditions of HAIR.
Given that aberrant accumulation of intracellular
iron induces oxidative stress (Galaris et al. 2019) and
subsequently results in multiple modes of cell death
(Leiet al. 2019), it is not surprising that, in addition to
ferroptosis, apoptosis (a non-inflammatory form of cell
death) and necroptosis (a pro-inflammatory form of cell
death) may also be involved in HAIR-induced fetal loss in
pregnant rats. Indeed, pregnant rats co-exposed to DHT
and INS exhibited decreased Casp3 mRNA expression and
cleaved caspase-3 protein abundance in the uterus, but
not in the placenta, even though selectively increased
expression of anti-apoptotic genes (Bcl-xl) and pro-
apoptotic genes (Bax) was observed in both tissues. We
suspect that suppression of apoptosis might serve as a
compensatory mechanism to protect against increased
ferroptosis in order to maintain homeostasis of the gravid
Downloaded from Bioscientifica.com at 10/31/2021 03:48:54PM
via free access
https://doi.org/10.1530/JOE-20-0155
https://joe.bioscientifica.com © 2020 Society for Endocrinology
Published by Bioscientifica Ltd.
Printed in Great Britain
260
Uterine and placental
ferroptosis in PCOS
Y Zhang, M Hu etal. 246:3
Journal of
Endocrinology
uterus after exposure to DHT and INS. This is supported by
studies assessing the interaction and interplay of different
cell death pathways in cancer research (Riegman et al.
2019). Ferroptosis and necroptosis are two different forms
of regulated necrosis (Xie et al. 2016, Choi et al. 2019).
Necroptosis requires mitochondrial ROS generation and
is primarily regulated by the RIPK1, RIPK3 and MLKL
proteins (Choiet al. 2019). We found that, in DHT+INS-
exposed pregnant rats, the level of ROS (Zhang et al.
2019b) and expression of Ripk1 and Ripk3 mRNAs was
increased in the placenta. Therefore, it is tempting to
speculate that the activation of necroptosis in response
to PCOS-related HAIR might serve to counteract the
ferroptosis pathway in the placenta. Of note, we also
found that maternal hyperandrogenism and insulin
resistance resulted in decreased ROS concentration in
the uterus ((Hu et al. 2019b) and this study). Given the
time-dependent regulation of uterine ROS levels in
normal rats during early-mid pregnancy (Huetal. 2019b),
it is possible that DHT + INS-induced ROS generation
and accumulation might not be sustained in the gravid
uterus on GD 14.5. Additionally, both ferroptosis and
necroptosis might intersect and crosstalk with HAIR-
induced oxidative damage and subsequently result in
increased fetal loss. It remains to be determined whether
HAIR-induced pregnancy loss is due to increased iron-
mediated uterine ferroptosis or to necroptosis-related
defects in the placenta, or both. There is evidence that
different forms of programmed cell death, including
ferroptosis, apoptosis, and necroptosis, may coexist under
the physiological condition and disease state (Choiet al.
2019, Riegmanetal. 2019). However, due to the limited
commercial antibodies available for rat tissues, we are
not able to extend the study to assess whether there is
co-existence of ferroptosis and apoptosis in the gravid
uterus and placenta in our experimental rats.
In this study, we found that some pro-ferroptosis genes
such as Acsl4, Tfrc and Dpp4 were oppositely regulated
in the uterus by co-exposure to DHT and INS. However,
several anti-ferroptosis genes, including Slc7a11, Gcls,
and Cisd1, were downregulated in the gravid uterus after
co-exposure to DHT and INS. These results suggest that
the suppression of anti-ferroptosis gene transcription
might play a dominant role in promoting ferroptosis
in this tissue under conditions of HAIR. Compared to
the gravid uterus, the placenta showed a distinct profile
of ferroptosis-related gene changes in response to the
combined DHT and INS exposure. For example, the
combined exposure increased Cisd1 mRNA expression
and decreased Dpp4 mRNA expression in the placenta,
which was opposite to that observed in the gravid
uterus. Furthermore, we often observed contrasting
expression patterns of pro- and anti-ferroptosis genes
in the gravid uterus and placenta with exposure to DHT
or INS alone compared to the combined exposure. We
do not know the exact reason for these inconsistencies;
however, we do know the ontogeny of changes we
observed and how these relate to the development of
HAIR as the expression of ferroptosis-related genes and
proteins was only assessed at one gestational age when
the pregnant rats already displayed HAIR (Hu et al.
2019b, Zhang et al. 2019b). In addition, components
of HAIR may have acted synergistically or through
separate pathways to bring about divergent effects on
gene expression and signaling pathways and regulate
the ferroptosis process in the gravid uterus and placenta.
Overall, our findings demonstrate the complexity and
challenges in establishing direct roles and patterns
linking individual pro-/anti-ferroptosis genes to the
ferroptosis pathway in the gravid uterus and placenta
in response to maternal DHT and/or INS in vivo. Future
work should therefore investigate the tissue-specific
and time-dependent changes in ferroptosis-related gene
expression in the uterus and placenta during the maternal
hormonal manipulation.
In comparison to the single exposure groups (DHT
or INS), specific changes within the maternal uterus and
placenta appeared to be driven by hyperandrogenism,
insulin resistance, or both (co-exposure to DHT and INS).
Experiments utilizing gene and pathway inhibitors in
decidual and trophoblast cells would be beneficial for
exploring the causality of changes observed regarding
ferroptosis and iron metabolism in the future. Work is
also required to assess whether elevated ferroptosis in
the uterus contributes to placental dysfunction in rat
dams with HAIR due to DHT and INS, a key area that
would additionally be aided by a time-course analysis.
Moreover, it is also possible that HAIR may activate
pathways within the uterus which serve to protect and
block the propagation of ferroptosis in placental tissue,
as opposed or in addition to intrinsic pathways operating
within the placenta itself. The gravid uterus and placenta
are composed of multiple cell types, each with their
distinct gene/protein expression program and likely
sensitivity to the DHT and INS treatment. Future work
should additionally undertake analyses of the ferroptosis
and apoptosis pathways in dissected placental zones or
isolated cell types from the gravid uterus and placenta
from rats treated with DHT and/or INS. Nonetheless, the
concomitant presence of different forms of regulated cell
Downloaded from Bioscientifica.com at 10/31/2021 03:48:54PM
via free access
https://doi.org/10.1530/JOE-20-0155
https://joe.bioscientifica.com © 2020 Society for Endocrinology
Published by Bioscientifica Ltd.
Printed in Great Britain
261
Research
Y Zhang, M Hu etal. Uterine and placental
ferroptosis in PCOS
246:3
Journal of
Endocrinology
death would be expected to disrupt uterine and placental
function and play a role in the fetal loss observed in
DHT + INS-exposed pregnant rats.
Recently, Zhang and colleagues reported that oxidative
stress-induced ferroptosis contributes to the pathogenesis
of preeclampsia (Zhang et al. 2020). In particular, they
found decreased GPX4, glutathione, and SLC7A11 protein
levels and increased MDA content in the preeclamptic
placenta in humans and rats (Zhangetal. 2020). Together
with our work, these findings support the notion that the
ferroptosis pathway is involved in the pathogenesis of
female reproductive disorders.
In summary, our findings suggest maternal
co-exposure to DHT and INS alters the ferroptosis pathway
in the gravid uterus and placenta; however, this occurs via
different regulatory mechanisms and signaling pathways.
For instance, in contrast to the placenta, increased
ferroptosis in the gravid uterus in response to DHT and
INS was related to decreased GPX4 and glutathione
abundance, altered expression of ferroptosis-associated
genes (Acsl4, Tfrc, Slc7a11, and Gclc), increased MDA
and iron deposition, upregulation of the ERK/p38/JNK
pathway and mitochondrial Dpp4 expression, as
well as the appearance of typical ferroptosis-related
mitochondrial morphology. In addition, DHT and INS
were associated with reduced activation of apoptosis in
the uterus and increased necroptosis in the placenta.
Both the maternal uterus and placenta play essential
roles in embryo implantation and support fetal growth
and development during pregnancy (Schatz et al. 2016,
Sharma et al. 2016). Therefore, while the present study
improves our understanding of the impact of HAIR on
regulated cell death in specific tissues during pregnancy,
more preclinical and clinical studies are needed to further
investigate the molecular and functional connectivity
between the maternal decidua, placenta and fetus in PCOS.
Supplementary materials
This is linked to the online version of the paper at https://doi.org/10.1530/
JOE-20-0155.
Declaration of interest
The authors declare that there is no conict of interest that could be
perceived as prejudicing the impartiality of the research reported.
Funding
This study was nanced by grants from the Swedish Medical Research
Council (grant number 10380), the Swedish state under the agreement
between the Swedish government and the county councils the ALF-
agreement (grant number ALFGBG-147791), Jane and Dan Olsson’s
Foundation, the Knut and Alice Wallenberg Foundation, and the Adlerbert
Research Foundation to HB and LRS as well as the National Natural Science
Foundation of China (Grant No. 81774136), the Project of Young Innovation
Talents in Heilongjiang Provincial University (Grant No.UNPYSCT-2015121),
the Scientic Research Foundation for Postdoctoral Researchers of Heilong
Jiang Province, the Project of Science Foundation by Heilongjiang University
of Chinese Medicine, and the Project of Excellent Innovation Talents by
Heilongjiang University of Chinese Medicine to YZ. The Guangzhou Medical
University High-level University Construction Talents Fund (grant number
B185006010046) supported MH. ANSP is supported by a Royal Society
Dorothy Hodgkin Research Fellowship.
Author contribution statement
LRS performed study design and supervision. YZ, MH, WJ, GL, JZ, BW, PC,
XL, YH, LS, XW, and LRS conducted the study. YZ, MH, WJ, GL, JZ, BW, JL, XL,
and LRS performed data collection. YZ, MH, JL, and LRS performed data
analysis. SL, ANSP, LS, MB, LRS, and HB performed data interpretation. YZ,
MH, and LRS drafted the manuscript. SL, ANSP, MB, LRS, and HB revised
the manuscript. YZ, MH, LRS, and HB take responsibility for the integrity of
the data analysis. All authors have read and approved the nal version of
the manuscript.
Acknowledgements
The funders had no role in the design, data collection, analysis, decision to
publish, or preparation of the manuscript.
References
AgarwalA, Aponte-MelladoA, PremkumarBJ, ShamanA & GuptaS
2012 The effects of oxidative stress on female reproduction: a
review. Reproductive Biology and Endocrinology 10 49. (https://doi.
org/10.1186/1477-7827-10-49)
AzzizR, CarminaE, ChenZ, DunaifA, LavenJS, LegroRS, LiznevaD,
Natterson-HorowtizB, TeedeHJ & YildizBO 2016 Polycystic ovary
syndrome. Nature Reviews: Disease Primers 2 16057. (https://doi.
org/10.1038/nrdp.2016.57)
Bahri KhomamiM, JohamAE, BoyleJA, PiltonenT, SilagyM,
AroraC, MissoML, TeedeHJ & MoranLJ 2019 Increased maternal
pregnancy complications in polycystic ovary syndrome appear to
be independent of obesity – a systematic review, meta-analysis,
and meta-regression. Obesity Reviews 20 659–674. (https://doi.
org/10.1111/obr.12829)
BaithaluRK, SinghSK, KumaresanA, MohantyAK, MohantyTK,
KumarS, KerkettaS, MaharanaBR, PatbandhaTK, AttupuramN,
etal. 2017 Transcriptional abundance of antioxidant enzymes in
endometrium and their circulating levels in Zebu cows with and
without uterine infection. Animal Reproduction Science 177 79–87.
(https://doi.org/10.1016/j.anireprosci.2016.12.008)
BanulsC, Rovira-LlopisS, Martinez de MaranonA, VesesS, JoverA,
GomezM, RochaM, Hernandez-MijaresA & VictorVM 2017
Metabolic syndrome enhances endoplasmic reticulum, oxidative
stress and leukocyte-endothelium interactions in PCOS. Metabolism:
Clinical and Experimental 71 153–162. (https://doi.org/10.1016/j.
metabol.2017.02.012)
CaoC & FlemingMD 2016 The placenta: the forgotten essential
organ of iron transport. Nutrition Reviews 74 421–431. (https://doi.
org/10.1093/nutrit/nuw009)
Downloaded from Bioscientifica.com at 10/31/2021 03:48:54PM
via free access
https://doi.org/10.1530/JOE-20-0155
https://joe.bioscientifica.com © 2020 Society for Endocrinology
Published by Bioscientifica Ltd.
Printed in Great Britain
262
Uterine and placental
ferroptosis in PCOS
Y Zhang, M Hu etal. 246:3
Journal of
Endocrinology
ChenL, HambrightWS, NaR & RanQ 2015 Ablation of the ferroptosis
inhibitor glutathione peroxidase 4 in neurons results in rapid motor
neuron degeneration and paralysis. Journal of Biological Chemistry 290
28097–28106. (https://doi.org/10.1074/jbc.M115.680090)
ChoiME, PriceDR, RyterSW & ChoiAMK 2019 Necroptosis: a crucial
pathogenic mediator of human disease. JCI Insight 4 e128834.
(https://doi.org/10.1172/jci.insight.128834)
ConradM, SchneiderM, SeilerA & BornkammGW 2007 Physiological
role of phospholipid hydroperoxide glutathione peroxidase in
mammals. Biological Chemistry 388 1019–1025. (https://doi.
org/10.1515/BC.2007.130)
DaiC, ChenX, LiJ, ComishP, KangR & TangD 2020 Transcription
factors in ferroptotic cell death. Cancer Gene Therapy [epub]. (https://
doi.org/10.1038/s41417-020-0170-2)
DaltoDB, RoyM, AudetI, PalinMF, GuayF, LapointeJ & MatteJJ 2015
Interaction between vitamin B6 and source of selenium on the
response of the selenium-dependent glutathione peroxidase system
to oxidative stress induced by oestrus in pubertal pig. Journal of Trace
Elements in Medicine and Biology 32 21–29. (https://doi.org/10.1016/j.
jtemb.2015.05.002)
DingY, JiangZ, XiaB, ZhangL, ZhangC & LengJ 2019 Mitochondria-
targeted antioxidant therapy for an animal model of PCOS-IR.
International Journal of Molecular Medicine 43 316–324. (https://doi.
org/10.3892/ijmm.2018.3977)
DixonSJ, LembergKM, LamprechtMR, SkoutaR, ZaitsevEM,
GleasonCE, PatelDN, BauerAJ, CantleyAM, YangWS, etal. 2012
Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell
149 1060–1072. (https://doi.org/10.1016/j.cell.2012.03.042)
Escobar-MorrealeHF 2012 Iron metabolism and the polycystic ovary
syndrome. Trends in Endocrinology & Metabolism 23 509–515. (https://
doi.org/10.1016/j.tem.2012.04.003)
FassnachtM, SchlenzN, SchneiderSB, WudySA, AllolioB & ArltW
2003 Beyond adrenal and ovarian androgen generation: increased
peripheral 5 alpha-reductase activity in women with polycystic ovary
syndrome. Journal of Clinical Endocrinology & Metabolism 88
2760–2766. (https://doi.org/10.1210/jc.2002-021875)
FengH, SchorppK, JinJ, YozwiakCE, HoffstromBG, DeckerAM,
RajbhandariP, StokesME, BenderHG, CsukaJM, etal. 2020
Transferrin receptor is a specific ferroptosis marker. Cell Reports 30
3411–3423.e7. (https://doi.org/10.1016/j.celrep.2020.02.049)
ForcinaGC & DixonSJ 2019 GPX4 at the crossroads of lipid homeostasis
and ferroptosis. Proteomics 19 e1800311. (https://doi.org/10.1002/
pmic.201800311)
Friedmann AngeliJP, SchneiderM, PronethB, TyurinaYY, TyurinVA,
HammondVJ, HerbachN, AichlerM, WalchA, EggenhoferE, etal.
2014 Inactivation of the ferroptosis regulator Gpx4 triggers acute
renal failure in mice. Nature Cell Biology 16 1180–1191. (https://doi.
org/10.1038/ncb3064)
GalarisD, BarboutiA & PantopoulosK 2019 Iron homeostasis and
oxidative stress: an intimate relationship. Biochimica et Biophysica
Acta. Molecular Cell Research 1866 118535. (https://doi.org/10.1016/j.
bbamcr.2019.118535)
GaoM, YiJ, ZhuJ, MinikesAM, MonianP, ThompsonCB & JiangX 2019
Role of mitochondria in ferroptosis. Molecular Cell 73 354–363.e3.
(https://doi.org/10.1016/j.molcel.2018.10.042)
GawelS, WardasM, NiedworokE & WardasP 2004 Malondialdehyde
(MDA) as a lipid peroxidation marker. Wiadomosci Lekarskie 57
453–455.
GlintborgD, JensenRC, BentsenK, SchmedesAV, BrandslundI, KyhlHB,
BilenbergN & AndersenMS 2018 Testosterone levels in third
trimester in polycystic ovary syndrome: odense child cohort. Journal
of Clinical Endocrinology & Metabolism 103 3819–3827. (https://doi.
org/10.1210/jc.2018-00889)
GudipatySA, ConnerCM, RosenblattJ & MontellDJ 2018
Unconventional ways to live and die: cell death and survival in
development, homeostasis, and disease. Annual Review of Cell &
Developmental Biology 34 311–332. (https://doi.org/10.1146/annurev-
cellbio-100616-060748)
GuoY, ZhangN, ZhangD, RenQ, GanzT, LiuS & NemethE 2019 Iron
homeostasis in pregnancy and spontaneous abortion. American
Journal of Hematology 94 184–188. (https://doi.org/10.1002/ajh.25341)
HuM, ZhangY, GuoX, JiaW, LiuG, ZhangJ, CuiP, LiJ, LiW, WuX,
etal. 2019a Perturbed ovarian and uterine glucocorticoid receptor
signaling accompanies the balanced regulation of mitochondrial
function and NFkappaB-mediated inflammation under conditions of
hyperandrogenism and insulin resistance. Life Sciences 232 116681.
(https://doi.org/10.1016/j.lfs.2019.116681)
HuM, ZhangY, GuoX, JiaW, LiuG, ZhangJ, LiJ, CuiP, Sferruzzi-
PerriAN, HanY, etal. 2019b Hyperandrogenism and insulin
resistance induce gravid uterine defects in association with
mitochondrial dysfunction and aberrant ROS production. American
Journal of Physiology – Endocrinology & Metabolism 316 E794–E809.
(https://doi.org/10.1152/ajpendo.00359.2018)
ImaiH, HiraoF, SakamotoT, SekineK, MizukuraY, SaitoM, KitamotoT,
HayasakaM, HanaokaK & NakagawaY 2003 Early embryonic
lethality caused by targeted disruption of the mouse PHGPx gene.
Biochemical & Biophysical Research Communications 305 278–286.
(https://doi.org/10.1016/s0006-291x(03)00734-4)
KimJW, KangKM, YoonTK, ShimSH & LeeWS 2014 Study of circulating
hepcidin in association with iron excess, metabolic syndrome, and
BMP-6 expression in granulosa cells in women with polycystic
ovary syndrome. Fertility & Sterility 102 548–554.e2. (https://doi.
org/10.1016/j.fertnstert.2014.04.031)
KimSJ, XiaoJ, WanJ, CohenP & YenK 2017 Mitochondrially derived
peptides as novel regulators of metabolism. Journal of Physiology 595
6613–6621. (https://doi.org/10.1113/JP274472)
KovtunovychG, EckhausMA, GhoshMC, Ollivierre-WilsonH &
RouaultTA 2010 Dysfunction of the heme recycling system in heme
oxygenase 1-deficient mice: effects on macrophage viability and tissue
iron distribution. Blood 116 6054–6062. (https://doi.org/10.1182/
blood-2010-03-272138)
LaiQ, XiangW, LiQ, ZhangH, LiY, ZhuG, XiongC & JinL 2018
Oxidative stress in granulosa cells contributes to poor oocyte quality
and IVF-ET outcomes in women with polycystic ovary syndrome.
Frontiers of Medicine 12 518–524. (https://doi.org/10.1007/s11684-017-
0575-y)
LeiP, BaiT & SunY 2019 Mechanisms of ferroptosis and relations with
regulated cell death: a review. Frontiers in Physiology 10 139. (https://
doi.org/10.3389/fphys.2019.00139)
LiJ, CaoF, YinHL, HuangZJ, LinZT, MaoN, SunB & WangG 2020
Ferroptosis: past, present and future. Cell Death & Disease 11 88.
(https://doi.org/10.1038/s41419-020-2298-2)
LiznevaD, SuturinaL, WalkerW, BraktaS, Gavrilova-JordanL & AzzizR
2016 Criteria, prevalence, and phenotypes of polycystic ovary
syndrome. Fertility & Sterility 106 6–15. (https://doi.org/10.1016/j.
fertnstert.2016.05.003)
LuSC 2009 Regulation of glutathione synthesis. Molecular Aspects of
Medicine 30 42–59. (https://doi.org/10.1016/j.mam.2008.05.005)
MaliqueoM, LaraHE, SanchezF, EchiburuB, CrisostoN & Sir-
PetermannT 2013 Placental steroidogenesis in pregnant women with
polycystic ovary syndrome. European Journal of Obstetrics, Gynecology,
and Reproductive Biology 166 151–155. (https://doi.org/10.1016/j.
ejogrb.2012.10.015)
MistryHD, KurlakLO, WilliamsPJ, RamsayMM, SymondsME &
Broughton PipkinF 2010 Differential expression and distribution
of placental glutathione peroxidases 1, 3 and 4 in normal and
preeclamptic pregnancy. Placenta 31 401–408. (https://doi.
org/10.1016/j.placenta.2010.02.011)
MistryHD, WilsonV, RamsayMM, SymondsME & Broughton PipkinF
2008 Reduced selenium concentrations and glutathione peroxidase
activity in preeclamptic pregnancies. Hypertension 52 881–888.
(https://doi.org/10.1161/HYPERTENSIONAHA.108.116103)
Downloaded from Bioscientifica.com at 10/31/2021 03:48:54PM
via free access
https://doi.org/10.1530/JOE-20-0155
https://joe.bioscientifica.com © 2020 Society for Endocrinology
Published by Bioscientifica Ltd.
Printed in Great Britain
263
Research
Y Zhang, M Hu etal. Uterine and placental
ferroptosis in PCOS
246:3
Journal of
Endocrinology
NgSW, NorwitzSG & NorwitzER 2019 The impact of iron overload
and ferroptosis on reproductive disorders in humans: implications
for preeclampsia. International Journal of Molecular Sciences 20 3283.
(https://doi.org/10.3390/ijms20133283)
PossKD & TonegawaS 1997 Heme oxygenase 1 is required for
mammalian iron reutilization. PNAS 94 10919–10924. (https://doi.
org/10.1073/pnas.94.20.10919)
RamosRS, OliveiraML, IzaguirryAP, VargasLM, SoaresMB, MesquitaFS,
SantosFW & BinelliM 2015 The periovulatory endocrine milieu
affects the uterine redox environment in beef cows. Reproductive
Biology and Endocrinology 13 39. (https://doi.org/10.1186/s12958-015-
0036-x)
RiegmanM, BradburyMS & OverholtzerM 2019 Population dynamics in
cell death: mechanisms of propagation. Trends in Cancer 5 558–568.
(https://doi.org/10.1016/j.trecan.2019.07.008)
RosenfieldRL & EhrmannDA 2016 The pathogenesis of polycystic ovary
syndrome (PCOS): the hypothesis of PCOS as functional ovarian
hyperandrogenism revisited. Endocrine Reviews 37 467–520. (https://
doi.org/10.1210/er.2015-1104)
SchatzF, Guzeloglu-KayisliO, ArlierS, KayisliUA & LockwoodCJ 2016
The role of decidual cells in uterine hemostasis, menstruation,
inflammation, adverse pregnancy outcomes and abnormal uterine
bleeding. Human Reproduction Update 22 497–515. (https://doi.
org/10.1093/humupd/dmw004)
SchneiderM, ForsterH, BoersmaA, SeilerA, WehnesH, SinowatzF,
NeumullerC, DeutschMJ, WalchA, Hrabe de AngelisM, etal. 2009
Mitochondrial glutathione peroxidase 4 disruption causes male
infertility. FASEB Journal 23 3233–3242. (https://doi.org/10.1096/
fj.09-132795)
SchootsMH, GordijnSJ, ScherjonSA, van GoorH & HillebrandsJL 2018
Oxidative stress in placental pathology. Placenta 69 153–161. (https://
doi.org/10.1016/j.placenta.2018.03.003)
SharmaS, GodboleG & ModiD 2016 Decidual control of trophoblast
invasion. American Journal of Reproductive Immunology 75 341–350.
(https://doi.org/10.1111/aji.12466)
SharpAN, HeazellAE, CrockerIP & MorG 2010 Placental apoptosis in
health and disease. American Journal of Reproductive Immunology 64
159–169. (https://doi.org/10.1111/j.1600-0897.2010.00837.x)
SilfenME, DenburgMR, ManiboAM, LoboRA, JaffeR, FerinM, LevineLS
& OberfieldSE 2003 Early endocrine, metabolic, and sonographic
characteristics of polycystic ovary syndrome (PCOS): comparison
between nonobese and obese adolescents. Journal of Clinical
Endocrinology & Metabolism 88 4682–4688. (https://doi.org/10.1210/
jc.2003-030617)
Sir-PetermannT, MaliqueoM, AngelB, LaraHE, Perez-BravoF &
RecabarrenSE 2002 Maternal serum androgens in pregnant women
with polycystic ovarian syndrome: possible implications in prenatal
androgenization. Human Reproduction 17 2573–2579. (https://doi.
org/10.1093/humrep/17.10.2573)
SpencerSJ, CataldoNA & JaffeRB 1996 Apoptosis in the human female
reproductive tract. Obstetrical & Gynecological Survey 51 314–323.
(https://doi.org/10.1097/00006254-199605000-00023)
StockwellBR, Friedmann AngeliJP, BayirH, BushAI, ConradM, DixonSJ,
FuldaS, GasconS, HatziosSK, KaganVE, etal. 2017 Ferroptosis: a
regulated cell death nexus linking metabolism, redox biology, and
disease. Cell 171 273–285. (https://doi.org/10.1016/j.cell.2017.09.021)
SunX, NiuX, ChenR, HeW, ChenD, KangR & TangD 2016
Metallothionein-1G facilitates sorafenib resistance through inhibition
of ferroptosis. Hepatology 64 488–500. (https://doi.org/10.1002/
hep.28574)
TangD, KangR, BergheTV, VandenabeeleP & KroemerG 2019 The
molecular machinery of regulated cell death. Cell Research 29
347–364. (https://doi.org/10.1038/s41422-019-0164-5)
WelshAO 1993 Uterine cell death during implantation and early
placentation. Microscopy Research & Technique 25 223–245. (https://
doi.org/10.1002/jemt.1070250305)
XieY, HouW, SongX, YuY, HuangJ, SunX, KangR & TangD 2016
Ferroptosis: process and function. Cell Death & Differentiation 23
369–379. (https://doi.org/10.1038/cdd.2015.158)
ZhangH, HeY, WangJX, ChenMH, XuJJ, JiangMH, FengYL & GuYF
2020 miR-30-5p-mediated ferroptosis of trophoblasts is implicated in
the pathogenesis of preeclampsia. Redox Biology 29 101402. (https://
doi.org/10.1016/j.redox.2019.101402)
ZhangJ, BaoY, ZhouX & ZhengL 2019a Polycystic ovary syndrome and
mitochondrial dysfunction. Reproductive Biology and Endocrinology:
RB&E 17 67. (https://doi.org/10.1186/s12958-019-0509-4)
ZhangY, MengF, SunX, SunX, HuM, CuiP, VestinE, LiX, LiW, WuXK,
etal. 2018 Hyperandrogenism and insulin resistance contribute to
hepatic steatosis and inflammation in female rat liver. Oncotarget 9
18180–18197. (https://doi.org/10.18632/oncotarget.24477)
ZhangY, SunX, SunX, MengF, HuM, LiX, LiW, WuXK, BrännströmM,
ShaoR, etal. 2016 Molecular characterization of insulin resistance
and glycolytic metabolism in the rat uterus. Scientific Reports 6 30679.
(https://doi.org/10.1038/srep30679)
ZhangY, ZhaoW, XuH, HuM, GuoX, JiaW, LiuG, LiJ, CuiP, LagerS,
etal. 2019 Hyperandrogenism and insulin resistance-induced fetal
loss: evidence for placental mitochondrial abnormalities and elevated
reactive oxygen species production in pregnant rats that mimic the
clinical features of polycystic ovary syndrome. Journal of Physiology
597 3927–3950. (https://doi.org/10.1113/JP277879)
ZhuLJ, BagchiMK & BagchiIC 1995 Ferritin heavy chain is a
progesterone-inducible marker in the uterus during pregnancy.
Endocrinology 136 4106–4115. (https://doi.org/10.1210/
endo.136.9.7649119)
Received in final form 28 May 2020
Accepted 25 June 2020
Accepted Manuscript published online 25 June 2020
Downloaded from Bioscientifica.com at 10/31/2021 03:48:54PM
via free access
... We also found upregulation of Nox2 by the placenta in HFHS mice, and NOX2 is implicated in the generation of ROS, is associated with ferroptosis [85] and is positively regulated by p38MAPK [86]. Prior work in a rat model of metabolic disease (insulin resistance) has also reported changes in iron deposition, activated ERK/p38/JNK and ferroptosis induction at the feto-maternal interface [87]. However, unlike that rat model of metabolic disease, which showed significantly augmented oxidative stress [87], the mRNA expression of anti-ferroptosis genes, including Gclc, Gss and Gpx4 [88] and other pro-ferroptosis genes, such as Acsl4 [89], were not significantly altered in the placentas from HFHS dietfed mice at either gestational age. ...
... Prior work in a rat model of metabolic disease (insulin resistance) has also reported changes in iron deposition, activated ERK/p38/JNK and ferroptosis induction at the feto-maternal interface [87]. However, unlike that rat model of metabolic disease, which showed significantly augmented oxidative stress [87], the mRNA expression of anti-ferroptosis genes, including Gclc, Gss and Gpx4 [88] and other pro-ferroptosis genes, such as Acsl4 [89], were not significantly altered in the placentas from HFHS dietfed mice at either gestational age. Therefore, it is possible that gestational changes in iron uptake by the placenta of HFHS diet-fed mice prevented a complete oxidative derailment, and in turn, a significant induction of ferroptosis. ...
Article
Full-text available
Obesity and gestational diabetes (GDM) impact fetal growth during pregnancy. Iron is an essential micronutrient needed for energy-intense feto-placental development, but if mis-handled can lead to oxidative stress and ferroptosis (iron-dependent cell death). In a mouse model showing maternal obesity and glucose intolerance, we investigated the association of materno-fetal iron handling and placental ferroptosis, oxidative damage and stress signalling activation with fetal growth. Female mice were fed a standard chow or high fat, high sugar (HFHS) diet during pregnancy and outcomes were measured at day (d)16 or d19 of pregnancy. In HFHS-fed mice, maternal hepcidin was reduced and iron status maintained (tissue iron levels) at both d16 and d19. However, fetal weight, placental iron transfer capacity, iron deposition, TFR1 expression and ERK2-mediated signalling were reduced and oxidative damage-related lipofuscin accumulation in the placenta was increased in HFHS-fed mice. At d19, whilst TFR1 remained decreased, fetal weight was normal and placental weight, iron content and iron transporter genes (Dmt1, Zip14, and Fpn1) were reduced in HFHS-fed mice. Furthermore, there was stress kinase activation (increased phosphorylated p38MAPK, total ERK and JNK) in the placenta from HFHS-fed mice at d19. In summary, a maternal HFHS diet during pregnancy impacts fetal growth trajectory in association with changes in placental iron handling, ferroptosis and stress signalling. Downregulation of placental iron transporters in HFHS mice may protect the fetus from excessive oxidative iron. These findings suggest a role for alterations in placental iron homeostasis in determining perinatal outcomes of pregnancies associated with GDM and/or maternal obesity. Graphical Abstract
... Briefly, rats in the PCOS group were subcutaneously injected daily with 60 mg/kg DHEA (MilliporeSigma) diluted with 0.2 ml sesame oil for 21 consecutive days. At the same time, the rats in the control group were injected with an equal volume of sesame oil (20)(21)(22)(23). Prior to DHEA injection to induce PCOS in rats, lentivirus carrying sh-TNC or sh-NC (10 µl; Shanghai GeneChem Co., Ltd.) was injected into the ovaries of rats. ...
Article
Polycystic ovary syndrome (PCOS) is a globally prevalent gynecological disorder among women of childbearing age. The present study aimed to investigate the role of tenascin C (TNC) in PCOS and its potential mechanisms. Fasting blood glucose and serum insulin, the homeostasis model assessment of insulin resistance and the serum hormone levels were determined in PCOS rats. In addition, H&E staining was used for assessing pathology. In addition, the effects of TNC on oxidative stress and inflammation response in PCOS rat and cell models was assessed. Furthermore, the roles of TNC on KGN cell proliferation and apoptosis were determined employing EdU assay and flow cytometry. TLR4/NF‑κB pathway‑related proteins were measured using western blotting, immunofluorescence and immunohistochemistry. It was found that the mRNA and protein expression was upregulated in PCOS rats and in KGN cells induced by dihydrotestosterone (DHT). Knockdown of TNC relieved the pathological characteristics and the endocrine abnormalities of PCOS rats. Knockdown of TNC inhibited ovarian cell apoptosis, oxidative stress and inflammation in PCOS rats. Knockdown of TNC reversed the DHT‑induced reduction in cell proliferation and increase in apoptosis in KGN cells. Furthermore, knockdown of TNC alleviated oxidative stress and inflammatory responses induced by DHT in KGN cells. Additionally, knockdown of TNC inhibited the toll‑like receptor 4 (TLR4)/NF‑κB signaling pathway in PCOS rats and DHT‑treated KGN cells. In conclusion, knockdown of TNC could ameliorate PCOS in both rats and a cell model by inhibiting cell apoptosis, oxidative stress and inflammation via the suppression of the TLR4/NF‑κB signaling pathway.
... Clinical manifestations of hyperandrogenism include hirsutism (excessive hair growth), androgenic acne, and/or alopecia (hair loss) brought on by high levels of androgen. Although there is not a method that is generally acknowledged for visually identifying androgenic acne, the Ludwig visual score can be used to determine the severity and pattern of alopecia [9][10][11]. The standard clinical measure for hirsutism assessment is the modified Ferriman Gallwey score (MFG), which is used in hirsutism diagnosis. ...
Article
Full-text available
Women in the reproductive age range are usually affected with Polycystic Ovary Syndrome (PCOS), a complex and multifaceted condition. Anovulation, hyperandrogenism, and metabolic difficulties like hyperglycemia, hypertension, and obesity in women are all manifestations of this condition, which also affects the reproductive system. The National Institutes of Health in the 1990s, Rotterdam in 2003, and Androgen Excess Polycystic Ovary Syndrome in 2009 all contributed to the evolution of the diagnostic criteria for PCOS. The 2003 Rotterdam criteria are currently the most generally used criteria. They call for at least two of the three criteria – irregular menstrual periods, polycystic ovary morphology on imaging, and hyperandrogenism – either clinically or biochemically – to be present in order to diagnose PCOS. It is currently being suggested that the anti-Müllerian hormone in serum be used instead of follicular count as an official indicator of polycystic ovarian morphology/PCOS. Hyperandrogenism and irregular periods are essential components in determining PCOS in adolescent patients. More recently, it has been shown that artificial intelligence, especially machine learning, holds great promise for detecting and predicting PCOS with high accuracy, potentially assisting in early management and treatment decisions. Examining the underlying mechanisms, clinical symptoms, and challenges involved in making a diagnosis of PCOS in females is the premise of this review article.
... Recent studies have revealed the aberrant expression of noncoding RNA in PCOS patients and then explored the potential effects of these noncoding RNAs on ferroptosis in immortal cell lines Tan et al., 2022). Abnormally expressed ferroptosis-related proteins in uterine and placental tissues in PCOS rat models have also been evaluated (Zhang et al., 2020c;Hu et al., 2021). No direct evidence has ever clarified the relative ferroptosis level in PCOS ovaries compared to the non-PCOS ovaries; therefore, the role of ferroptosis in the pathogenesis of PCOS and the underlying mechanism involved remained unknown. ...
Article
STUDY QUESTION Does ovarian ferroptosis play an active role in the development of polycystic ovary syndrome (PCOS)? SUMMARY ANSWER Increased ovarian ferroptosis was present in PCOS ovaries and the inhibition of ferroptosis with ferrostatin-1 (Fer-1) ameliorated polycystic ovary morphology and anovulation. WHAT IS KNOWN ALREADY Programmed cell death plays a fundamental role in ovarian follicle development. However, the types and mechanisms of cell death involved in the ovary are yet to be elucidated. Ferroptosis is a recently discovered iron-dependent programmed cell death. Impaired iron metabolism and cell death have been observed in women with PCOS, the main cause of anovulatory infertility. Additionally, previous studies reported that an abnormal expression of noncoding RNA may promote ferroptosis in immortalized ovarian granulosa cell lines. However, little is known about whether ovarian ferroptosis is increased in PCOS, and there is insufficient direct evidence for a role of ferroptosis in PCOS, and the underlying mechanism. Moreover, the effect of the inhibition of ferroptosis with Fer-1 in PCOS remains unclear. STUDY DESIGN, SIZE, DURATION Ferroptosis was evaluated in human granulosa cells (hGCs) from non-PCOS (n = 6–16) and PCOS (n = 7–18) patients. The experimental study was completed in vitro using primary hGCs from women undergoing IVF. Improvements in PCOS indicators following ferroptosis inhibition with Fer-1 were investigated in a dehydroepiandrosterone (DHEA)-induced PCOS rat model (n = 8 per group). PARTICIPANTS/MATERIALS, SETTING, METHODS Ovarian ferroptosis was evaluated in the following ways: by detecting iron concentrations via ELISA and fluorescent probes; measuring malondialdehyde (MDA) concentrations via ELISA; assessing ferroptosis-related protein abundance with western blotting; observing mitochondrial morphology with transmission electron microscopy; and determining cell viability. Primary hGCs were collected from women undergoing IVF. They were treated with dihydrotestosterone (DHT) for 24 h. The effect of DHT on ferroptosis was examined in the presence or absence of small interfering RNA-mediated knockdown of the putative receptor coregulator for signaling molecules. The role of ovarian ferroptosis in PCOS progression was explored in vivo in rats. The DHEA-induced PCOS rat model was treated with the ferroptosis inhibitor, Fer-1, and the oocytes and metaphase II oocytes were counted after ovarian stimulation. Additionally, rats were treated with the ferroptosis inducer, RSL3, to further explore the effect of ferroptosis. The concentrations of testosterone, FSH, and LH were assessed. MAIN RESULTS AND THE ROLE OF CHANCE Increased ferroptosis was detected in the ovaries of patients with PCOS and in rats with DHEA-induced PCOS. Increased concentrations of Fe2+ (P < 0.05) and MDA (P < 0.05), and upregulated nuclear receptor coactivator 4 protein levels, and downregulated ferritin heavy chain 1 (FTH1) and glutathione peroxidase 4 (GPX4) proteins were observed in the hGCs in patients with PCOS and ovaries of PCOS rats (P < 0.05 versus control). DHT was shown to induce ferroptosis via activation of NOCA4-dependent ferritinophagy. The inhibition of ferroptosis with Fer-1 in rats ameliorated a cluster of PCOS traits including impaired glucose tolerance, irregular estrous cycles, reproductive hormone dysfunction, hyperandrogenism, polycystic ovaries, anovulation, and oocyte quality (P < 0.05). Treating rats with RSL3 resulted in polycystic ovaries and hyperandrogenism (P < 0.05). LARGE-SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Although ovarian-targeted ferroptosis inhibition may be a more targeted treatment for PCOS, the underlying mechanisms in the cycle between ferroptosis and hyperandrogenism require further exploration. Additionally, since PCOS shows high heterogeneity, it is important to investigate whether ferroptosis increases are present in all patients with PCOS. WIDER IMPLICATIONS OF THE FINDINGS Androgen-induced ovarian ferroptosis appears to play a role in the pathogenesis of PCOS, which potentially makes it a promising treatment target in PCOS. STUDY FUNDING/COMPETING INTEREST(S) This study was supported by the National Key R&D Program of China (2023YFC2705500, 2023YFC2705505, 2019YFA0802604), National Natural Science Foundation of China (No. 82130046, 82320108009, 82101708, 82101747, and 82001517), Shanghai leading talent program, Innovative research team of high-level local universities in Shanghai (No. SHSMU-ZLCX20210201, No. SSMU-ZLCX20180401), Shanghai Jiaotong University School of Medicine, Affiliated Renji Hospital Clinical Research Innovation Cultivation Fund Program (RJPY-DZX-003) and Shanghai Municipal Education Commission—Gaofeng Clinical Medicine Grant Support (No. 20161413), Shanghai’s Top Priority Research Center Construction Project (2023ZZ02002), and Three-Year Action Plan for Strengthening the Construction of the Public Health System in Shanghai (GWVI-11.1-36). The authors report no competing interests.
... Second, studies on PCOS-like rats indicate that IR causes the activation of ferroptosis in the gravid uterus and placenta. Furthermore, necroptosis and apoptosis might play a role in compensating or coordinating for IR-induced ferroptosis when the gravid uterine and placental dysfunction occur (33). In addition, hyperinsulinemia and IR can lead to increased secretion of reactive oxygen species, which further cause mitochondrial and placental dysfunction after pregnancy, thus increasing the risk of miscarriage and decreasing the live birth rate (34,35). ...
Article
Full-text available
Objective To investigate the effects of β-cell dysfunction on IVF outcomes in women with PCOS. Methods This retrospective cohort study includes 1,212 women with PCOS undergoing their first IVF cycle between September 2010 and December 2019. Beta-cell dysfunction was measured by homeostasis model assessment of β-cell function (HOMA-β) index. Results In quartiles of HOMA-β, the incidence of miscarriage dramatically increased from 10.2% (Q1) to 31.1% (Q4) (P for trend <0.001). Likewise, the incidence of miscarriage in quartiles of HOMA-β also showed a similar trend (P for trend <0.001). After adjusting for confounding factors, logistic regression analyses showed that high HOMA-IR values were independently associated with a high risk of miscarriage, with the odds ratios (OR) and 95% confidence intervals for quartiles 2–4 versus quartile 1 were 1.30 (0.69-2.46), 1.82 (0.97-3.43), and 3.57 (1.86-6.85), respectively (P for trend <0.001). When analyzed jointly, women in the highest HOMA-IR and highest HOMA-β group exhibited the highest risk for miscarriage compared with all other groups. Furthermore, higher HOMA-IR values were associated with higher risks of miscarriage among PCOS women regardless of HOMA-β values. Conclusions β-cell dysfunction is independently associated with increased miscarriage rate and decreased live birth rate in women with PCOS. It also plays a synergistic role with IR in terms of the reproductive outcomes, while the influence of IR overweighs that of β-cell dysfunction.
Article
Objective Polycystic ovary syndrome (PCOS) is the most prevalent endocrine disorder in women of reproductive age. The objective of our investigation is to study the protective effect of hydroethanolic extract of palm meristem (HEPM) on female rats with PCOS. Materials and Methods The experimental study involved the placement of 42 adult female Wistar rats ( n = 7) into six groups, with the control group receiving 0.25 ml of physiological serum by gavage daily, the sham group getting 0.25 ml of olive oil, and the HEPM 250, 500, and 500 per se group taking HEPM by gavage daily. PCOS was induced with estradiol valerate (EV), which was administered through a subcutaneous injection at 2 mg/kg volume using 0.25 ml of olive oil. After experiments, all animals were subjected to anesthesia, and serums were extracted for biochemical and hormonal evaluation. Results EV showed marked increases in serum levels of testosterone and luteinizing hormone when compared with the control group. Significantly, the EV group decreased 17-β estradiol, progesterone, and follicle-stimulating hormone. There was a significant alteration in hormonal levels across the treatment groups. The serum malondialdehyde levels and total oxidant status were elevated in the EV group. In addition, the serum levels of total antioxidant capacity, glutathione, and glutathione peroxidase were significantly lower in the EV group than in control rats. In the treatment groups, HEPM significantly reverses the oxidant/antioxidant balance and ameliorates the adverse effect of PCOS on oxidative stress. Conclusion The study findings indicated that the HEPM effectively safeguarded ovarian tissue from developing PCOS in the presence of EV.
Article
Baicalin has been proven to have the potential to reduce apoptosis and diabetic cardiomyopathy (DCM). However, the mechanism behind this effect still needs to be fully understood. To explore the potential therapeutic properties of Baicalin in managing DCM and controlling glycemic levels. In this study, Baicalin (at doses of 20, 60, or 120 mg/kg/d) were used to treat diabetic rats. At the end of treatment, the heart function of the rats was assessed. Furthermore, their serum levels of TG, TC, and LDL were measured using the ELISA method. Cell viability was evaluated using the CCK8 assay and apoptosis was assessed using flow cytometry or TUNEL assay. Primary cardiomyocytes were infected with NRF2 siRNA and then treated with Baicalin while incubating with high glucose (25 mmol/L). Protein and mRNA variations were analyzed using Western blot and qRT-PCR, respectively. The study found that when given Baicalin, diabetic rats demonstrated improved heart function. Without treatment, the hearts of diabetic rats displayed elevated levels of apoptotic cell death and cardiomyocyte autophagy, as well as decreased expressions of NRF2, HO-1, and KEAP1. However, Baicalin was able to reverse all of these diabetes-induced biochemical changes. Treatment enhanced NRF2 nuclear transfer, reduced hyperglycemia-induced apoptosis and autophagy in primary cardiomyocytes, and improved cellular viability in in vitro experiments. It must be noted that the protective effects of Baicalin were only observed when the Nrf2 gene expression was present in primary cardiomyocytes. Baicalin may reduce the effects of DCM by activating NRF2 through KEAP1 suppression and regulating autophagy activation.
Article
Full-text available
The discovery of mitochondria-derived peptides has facilitated a comprehensive understanding of their protective effects on various organs. One of such peptides, Mitochondrial ORF of the 12S rRNA type-C (MOTS-c), was initially characterized in 2015 as a bioactive molecule that regulates gene expression and cellular metabolism via 5’-adenosine monophosphate-activated protein kinase (AMPK). MOTS-c has exhibited notable protective effects across diverse organs, including protection against diabetes, cardiovascular diseases, alleviating the impacts of ageing, and regulating the immune response. Despite these well-established functions, the precise role of MOTS-c in the endocrine system remains elusive. However, recent research emphasizes the increasing significance of MOTS-c and other mitochondrial-derived peptides in regulating endocrine system function and addressing metabolism-related diseases. Therefore, this review aims to summarize the current information on the action of MOTS-c and other mitochondrial--derived peptides in various endocrine system organs.
Article
Full-text available
Ferroptosis is a type of regulated cell death driven by the iron-dependent accumulation of oxidized polyunsaturated fatty acid-containing phospholipids. There is no reliable way to selectively stain ferroptotic cells in tissue sections to characterize the extent of ferroptosis in animal models or patient samples. We address this gap by immunizing mice with membranes from lymphoma cells treated with the ferroptosis inducer piperazine erastin and screening ∼4,750 of the resulting monoclonal antibodies generated for their ability to selectively detect cells undergoing ferroptosis. We find that one antibody, 3F3 ferroptotic membrane antibody (3F3-FMA), is effective as a selective ferroptosis-staining reagent. The antigen of 3F3-FMA is identified as the human transferrin receptor 1 protein (TfR1). We validate this finding with several additional anti-TfR1 antibodies and compare them to other potential ferroptosis-detecting reagents. We find that anti-TfR1 and anti-malondialdehyde adduct antibodies are effective at staining ferroptotic tumor cells in multiple cell culture and tissue contexts.
Article
Full-text available
Oxidative stress is a major cause of adverse outcomes in preeclampsia (PE). Ferroptosis, i.e. programmed cell death from iron-dependent lipid peroxidation, likely mediates PE pathogenesis. We evaluated specific markers for ferroptosis in normal and PE placental tissues, using in vitro (trophoblasts) and in vivo (rat) models. Increase in malondialdehyde content and total Fe2+ along with reduced the glutathione content and glutathione peroxidase activity was observed in PE placenta. While the trophoblasts experienced death under hypoxia, inhibitors of ferroptosis, apoptosis, autophagy, and necrosis increased the cell viability. Microarrays, bioinformatic analysis, and luciferase reporter assay revealed that upregulation of miR-30b-5p in PE models plays a pivotal role in ferroptosis, by downregulating Cys2/glutamate antiporter and PAX3 and decreasing ferroportin 1 (an iron exporter) expression, resulting in decreased GSH and increased labile Fe2+. Inhibition of miR-30b-5p expression and supplementation with ferroptosis inhibitors attenuated the PE symptoms in rat models, making miR-30b-5p a potential therapeutic target for PE. Keywords: miR-30-5p, Ferroptosis, Preeclampsia, SLC7A11, Ferroportin 1
Article
Full-text available
Abstract Polycystic ovary syndrome (PCOS) is a prevalent hormonal disorder of premenopausal women worldwide and is characterized by reproductive, endocrine, and metabolic abnormalities. The clinical manifestations of PCOS include oligomenorrhea or amenorrhea, hyperandrogenism, ovarian polycystic changes, and infertility. Women with PCOS are at an increased risk of suffering from type 2 diabetes; me\tabolic syndrome; cardiovascular events, such as hypertension, dyslipidemia; gynecological diseases, including infertility, endometrial dysplasia, endometrial cancer, and ovarian malignant tumors; pregnancy complications, such as premature birth, low birthweight, and eclampsia; and emotional and mental disorders in the future. Although numerous studies have focused on PCOS, the underlying pathophysiological mechanisms of this disease remain unclear. Mitochondria play a key role in energy production, and mitochondrial dysfunction at the cellular level can affect systemic metabolic balance. The recent wide acceptance of functional mitochondrial disorders as a correlated factor of numerous diseases has led to the presupposition that abnormal mitochondrial metabolic markers are associated with PCOS. Studies conducted in the past few years have confirmed that increased oxidative stress is associated with the progression and related complications of PCOS and have proven the relationship between other mitochondrial dysfunctions and PCOS. Thus, this review aims to summarize and discuss previous and recent findings concerning the relationship between mitochondrial dysfunction and PCOS.
Article
Full-text available
Iron is an essential element for the survival of most organisms, including humans. Demand for iron increases significantly during pregnancy to support growth and development of the fetus. Paradoxically, epidemiologic studies have shown that excessive iron intake and/or high iron status can be detrimental to pregnancy and is associated with reproductive disorders ranging from endometriosis to preeclampsia. Reproductive complications resulting from iron deficiency have been reviewed elsewhere. Here, we focus on reproductive disorders associated with iron overload and the contribution of ferroptosis—programmed cell death mediated by iron-dependent lipid peroxidation within cell membranes—using preeclampsia as a model system. We propose that the clinical expressions of many reproductive disorders and pregnancy complications may be due to an underlying ferroptopathy (elemental iron-associated disease), characterized by a dysregulation in iron homeostasis leading to excessive ferroptosis.
Article
Full-text available
Key points Women with polycystic ovary syndrome (PCOS) commonly suffer from miscarriage, but the underlying mechanisms remain unknown. Herein, pregnant rats chronically treated with 5α‐dihydrotestosterone (DHT) and insulin exhibited hyperandrogenism and insulin resistance, as well as increased fetal loss, and these features are strikingly similar to those observed in pregnant PCOS patients. Fetal loss in our DHT+insulin‐treated pregnant rats was associated with mitochondrial dysfunction, disturbed superoxide dismutase 1 and Keap1/Nrf2 antioxidant responses, over‐production of reactive oxygen species (ROS) and impaired formation of the placenta. Chronic treatment of pregnant rats with DHT or insulin alone indicated that DHT triggered many of the molecular pathways leading to placental abnormalities and fetal loss, whereas insulin often exerted distinct effects on placental gene expression compared to co‐treatment with DHT and insulin. Treatment of DHT+insulin‐treated pregnant rats with the antioxidant N‐acetylcysteine improved fetal survival but was deleterious in normal pregnant rats. Our results provide insight into the fetal loss associated with hyperandrogenism and insulin resistance in women and suggest that physiological levels of ROS are required for normal placental formation and fetal survival during pregnancy. Abstract Women with polycystic ovary syndrome (PCOS) commonly suffer from miscarriage, but the underlying mechanism of PCOS‐induced fetal loss during pregnancy remains obscure and specific therapies are lacking. We used pregnant rats treated with 5α‐dihydrotestosterone (DHT) and insulin to investigate the impact of hyperandrogenism and insulin resistance on fetal survival and to determine the molecular link between PCOS conditions and placental dysfunction during pregnancy. Our study shows that pregnant rats chronically treated with a combination of DHT and insulin exhibited endocrine aberrations such as hyperandrogenism and insulin resistance that are strikingly similar to those in pregnant PCOS patients. Of pathophysiological significance, DHT+insulin‐treated pregnant rats had greater fetal loss and subsequently decreased litter sizes compared to normal pregnant rats. This negative effect was accompanied by impaired trophoblast differentiation, increased glycogen accumulation, and decreased angiogenesis in the placenta. Mechanistically, we report that over‐production of reactive oxygen species (ROS) in the placenta, mitochondrial dysfunction, and disturbed superoxide dismutase 1 (SOD1) and Keap1/Nrf2 antioxidant responses constitute important contributors to fetal loss in DHT+insulin‐treated pregnant rats. Many of the molecular pathways leading to placental abnormalities and fetal loss in DHT+insulin treatment were also seen in pregnant rats treated with DHT alone, whereas pregnant rats treated with insulin alone often exerted distinct effects on placental gene expression compared to insulin treatment in combination with DHT. We also found that treatment with the antioxidant N‐acetylcysteine (NAC) improved fetal survival in DHT+insulin‐treated pregnant rats, an effect related to changes in Keap1/Nrf2 and nuclear factor‐κB signalling. However, NAC administration resulted in fetal loss in normal pregnant rats, most likely due to PCOS‐like endocrine abnormality induced by the treatment. Our results suggest that the deleterious effects of hyperandrogenism and insulin resistance on fetal survival are related to a constellation of mitochondria–ROS–SOD1/Nrf2 changes in the placenta. Our findings also suggest that physiological levels of ROS are required for normal placental formation and fetal survival during pregnancy.
Article
Ferroptosis, a form of regulated cell death, is characterized by an excessive degree of iron accumulation and lipid peroxidation. Although it was originally identified only in cells expressing a mutant RAS oncogene, ferroptosis has also been found in normal cells following treatment by small molecules (e.g., erastin and RSL3) or drugs (e.g., sulfasalazine, sorafenib, and artesunate), which target antioxidant enzyme systems, especially the amino acid antiporter system xc− and the glutathione peroxidase GPX4. Dysfunctional ferroptosis is implicated in various physiological and pathological processes (e.g., metabolism, differentiation, and immunity). Targeting the ferroptotic network appears to a new treatment option for diseases or pathological conditions (e.g., cancer, neurodegeneration, and ischemia reperfusion injury). While the molecular machinery of ferroptosis remains largely unknown, several transcription factors (e.g., TP53, NFE2L2/NRF2, ATF3, ATF4, YAP1, TAZ, TFAP2C, SP1, HIF1A, EPAS1/HIF2A, BACH1, TFEB, JUN, HIC1, and HNF4A) play multiple roles in shaping ferroptosis sensitivity through either transcription-dependent or transcription-independent mechanisms. In this review, we summarize recent progress in understanding the transcriptional regulation underlying ferroptotic cell death, and discuss how it has provided new insights into cancer therapy.
Article
Iron is a transition metal and essential constituent of almost all living cells and organisms. As component of various metalloproteins it is involved in critical biochemical processes such as transport of oxygen in tissues, electron transfer reactions during respiration in mitochondria, synthesis and repair of DNA, metabolism of xenobiotics, etc. However, when present in excess within cells and tissues, iron disrupts redox homeostasis and catalyzes the propagation of reactive oxygen species (ROS), leading to oxidative stress. ROS are critical for physiological signaling pathways, but oxidative stress is associated with tissue injury and disease. At the cellular level, oxidative stress may lead to ferroptosis, an iron-dependent form of cell death. In this review, we focus on the intimate relationship between iron metabolism and oxidative stress in health and disease. We discuss aspects of redox- and iron-mediated signaling, toxicity, ferroptotic cell death, homeostatic pathways and pathophysiological implications.
Article
Cell death can occur through numerous regulated mechanisms that are categorized by their molecular machineries and differing effects on physiology. Apoptosis and necrosis, for example, have opposite effects on tissue inflammation due to their different modes of execution. Another feature that can distinguish different forms of cell death is that they have distinct intrinsic effects on the cell populations in which they occur. For example, a regulated mechanism of necrosis called ferroptosis has the unusual ability to spread between cells in a wave-like manner, thereby eliminating entire cell populations. Here we discuss the ways in which cell death can propagate between cells in normal physiology and disease, as well as the potential exploitation of cell death propagation for cancer therapy.
Article
Necroptosis is a genetically regulated form of necrotic cell death that has emerged as an important pathway in human disease. The necroptosis pathway is induced by a variety of signals, including death receptor ligands, and regulated by receptor-interacting protein kinases 1 and 3 (RIPK1 and RIPK3) and mixed-lineage kinase domain-like pseudokinase (MLKL), which form a regulatory necrosome complex. RIPK3-mediated phosphorylation of MLKL executes necroptosis. Recent studies, using animal models of tissue injury, have revealed that RIPK3 and MLKL are key effectors of injury propagation. This Review explores the functional roles of RIPK3 and MLKL as crucial pathogenic determinants and markers of disease progression and severity in experimental models of human disease, including acute and chronic pulmonary diseases; renal, hepatic, cardiovascular, and neurodegenerative diseases; cancer; and critical illness.
Article
Aim This study aimed to determine whether glucocorticoid receptor (GR) signaling, mitochondrial function, and local inflammation in the ovary and uterus are intrinsically different in rats with hyperandrogenism and insulin resistance compared to controls. Main methods Female Sprague Dawley rats were exposed to daily injections of human chorionic gonadotropin and/or insulin. Key findings In both the ovary and the uterus, decreased expression of the two GR isoforms was concurrent with increased expression of Fkbp51 but not Fkbp52 mRNA in hCG + insulin-treated rats. However, these rats exhibited contrasting regulation of Hsd11b1 and Hsd11b2 mRNAs in the two tissues. Further, the expression of several oxidative phosphorylation-related proteins decreased in the ovary and uterus following hCG and insulin stimulation, in contrast to increased expression of many genes involved in mitochondrial function and homeostasis. Additionally, hCG + insulin-treated rats showed increased expression of ovarian and uterine NFκB signaling proteins and Tnfaip3 mRNA. The mRNA expression of Il1b, Il6, and Mmp2 was decreased in both tissues, while the mRNA expression of Tnfa, Ccl2, Ccl5, and Mmp3 was increased in the uterus. Ovaries and uteri from animals co-treated with hCG and insulin showed increased collagen deposition compared to controls. Significance Our observations suggest that hyperandrogenism and insulin resistance disrupt ovarian and uterine GR activation and trigger compensatory or adaptive effects for mitochondrial homeostasis, allowing tissue-level maintenance of mitochondrial function in order to limit ovarian and uterine dysfunction. Our study also suggests that hyperandrogenism and insulin resistance activate NFκB signaling resulting in aberrant regulation of inflammation-related gene expression. Keywords Glucocorticoid receptor11HSDMitochondrial malfunctionNFκBInflammationOvaryUterusPolycystic ovary syndrome