ArticlePDF Available

Effects of Bisphenol A on reproductive toxicity and gut microbiota dysbiosis in male rats

Authors:

Abstract and Figures

Bisphenol A (BPA) is an environmental endocrine disruptor. Recent studies have shown an association between decreased spermatogenesis and gut microbiota alteration. However, the potential associations and mechanisms of BPA exposure on spermatogenesis, hormone production, and gut microbiota remain unknown. This study aims to investigate BPA-induced male reproductive toxicity and the potential link with gut microbiota dysbiosis. Male Sprague Dawley rats were exposed to BPA at different doses by oral gavage for thirty consecutive days. The extent of testicular damage was evaluated by basic parameters of body weight and hematoxylin-eosin (H&E) staining. Next, we determined the mRNA levels and protein levels of apoptosis, histone-related factors, and mammalian target of rapamycin (mTOR) pathway in testes. Finally, 16 S rDNA sequencing was used to analyze gut microbiota composition after BPA exposure. BPA exposure damaged testicular histology, significantly decreased sperm count, and increased sperm abnormalities. In addition, BPA exposure caused oxidative stress and cell apoptosis in testes. The levels of histone (H2A, H3) were significantly increased, while ubiquitin histone H2A (ub-H2A) and ubiquitin histone H2B (ub-H2B) were markedly reduced. Furthermore, BPA activated the PI3K and AKT expression, but the protein expressions of mTOR and 4EBP1 in testes were inhibited significantly. Additionally, the relative abundance of class Gammaproteobacteria, and order Betaproteobacteriales was significantly higher when treated with a high dose of BPA compared to the control group, which was negatively correlated with testosterone level. This study highlights the relationship between BPA-induced reproductive toxicity and gut microbiota disorder and provides new insights into the prevention and treatment of BPA-induced reproductive damage.
Content may be subject to copyright.
Ecotoxicology and Environmental Safety 239 (2022) 113623
Available online 11 May 2022
0147-6513/© 2022 Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
Effects of Bisphenol A on reproductive toxicity and gut microbiota dysbiosis
in male rats
Ruijing Liu
a
, Dongbao Cai
b
, Xusheng Li
b
, Boping Liu
a
, Jiali Chen
b
, Xinwei Jiang
b
, Haiwei Li
b
,
Zhenhua Li
c
, Katja Teerds
d
, Jianxia Sun
e
, Weibin Bai
b
,
*
, Yulong Jin
a
,
**
a
Key Laboratory for Bio-Based Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou
510630, PR China
b
Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid
Detection, Jinan University, Guangzhou 510632, PR China
c
Zhuhai Precision Medical Center, Zhuhai Peoples Hospital (Zhuhai Hospital Afliated with Jinan University), Jinan University, Zhuhai 519070, PR China
d
Department of Animal Sciences, Human and Animal Physiology, Wageningen University, Wageningen, The Netherlands
e
School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, PR China
ARTICLE INFO
Edited by Dr Yong Liang
Keywords:
Bisphenol A
Male reproduction
Hormone
Mammalian target of rapamycin
Apoptosis
Gut microbiota
ABSTRACT
Bisphenol A (BPA) is an environmental endocrine disruptor. Recent studies have shown an association between
decreased spermatogenesis and gut microbiota alteration. However, the potential associations and mechanisms
of BPA exposure on spermatogenesis, hormone production, and gut microbiota remain unknown. This study aims
to investigate BPA-induced male reproductive toxicity and the potential link with gut microbiota dysbiosis. Male
Sprague Dawley rats were exposed to BPA at different doses by oral gavage for thirty consecutive days. The
extent of testicular damage was evaluated by basic parameters of body weight and hematoxylin-eosin (H&E)
staining. Next, we determined the mRNA levels and protein levels of apoptosis, histone-related factors, and
mammalian target of rapamycin (mTOR) pathway in testes. Finally, 16 S rDNA sequencing was used to analyze
gut microbiota composition after BPA exposure. BPA exposure damaged testicular histology, signicantly
decreased sperm count, and increased sperm abnormalities. In addition, BPA exposure caused oxidative stress
and cell apoptosis in testes. The levels of histone (H2A, H3) were signicantly increased, while ubiquitin histone
H2A (ub-H2A) and ubiquitin histone H2B (ub-H2B) were markedly reduced. Furthermore, BPA activated the
PI3K and AKT expression, but the protein expressions of mTOR and 4EBP1 in testes were inhibited signicantly.
Additionally, the relative abundance of class Gammaproteobacteria, and order Betaproteobacteriales was
signicantly higher when treated with a high dose of BPA compared to the control group, which was negatively
correlated with testosterone level. This study highlights the relationship between BPA-induced reproductive
toxicity and gut microbiota disorder and provides new insights into the prevention and treatment of BPA-induced
reproductive damage.
Abbreviations: BPA, Bisphenol A; NOAEL, No observed adverse effect level; LOAEL, Lowest observed adverse effect level; PI3K, Phosphoinositide 3-kinase; mTOR,
Mammalian target of rapamycin; 4EBP1, 4E-binding protein 1; EDC, Endocrine-disrupting chemical; LH, Luteinizing hormone; FSH, Follicle-stimulating hormone;
DHT, Dihydrotestosterone; INHB, Inhibin B; E2, Estradiol; GnRH, Gonadotrophin-releasing hormone; HPG, Hypothalamic-pituitary-gonadal; EE, Ethinyl estradiol;
Bax, Bcl-2-associated X protein; Bcl-2, B-cell lymphoma 2; TUNEL, Terminal deoxynucleotidyl transferase-mediated nick end labeling; eIF4E, Eukaryotic translation
initiation factor 4E; PARP, Poly (ADP-ribose) polymerase; Raptor, Regulatory-associated protein of mTOR; SOD, Superoxide dismutase; GSH, Glutathione; CAT,
Catalase; MDA, Malondialdehyde; GPR54, G protein-coupled receptor 54; BTB, Blood-testis barrier.
* Correspondence to: Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Jinan University, 601 Huangpu Rd, Guangzhou 510632,
PR China.
** Corresponding author.
E-mail addresses: baiweibin@163.com (W. Bai), jyl@scau.edu.cn (Y. Jin).
Contents lists available at ScienceDirect
Ecotoxicology and Environmental Safety
journal homepage: www.elsevier.com/locate/ecoenv
https://doi.org/10.1016/j.ecoenv.2022.113623
Received 12 March 2022; Received in revised form 23 April 2022; Accepted 7 May 2022
Ecotoxicology and Environmental Safety 239 (2022) 113623
2
1. Introduction
Bisphenol A (BPA), a known endocrine-disrupting chemical (EDC)
with estrogenic activity, is a synthetic chemical that is found in large
quantities in the environment (Liu et al., 2021b; Molina et al., 2018). It
is widely used to produce polycarbonate plastics, epoxy resin liners for
food cans, water bottles, some dental sealants and composites, thermal
receipts, and other applications (Ehrlich et al., 2014). BPA can migrate
into the water and food under conditions such as high temperature,
acidity, and alkalinity (Vandenberg et al., 2009). The routes of human
exposure to BPA are various including inhalation (dust, occupational
sources) and dermal contact (household products, cosmetics, medical
devices) (Cho et al., 2012; Demierre et al., 2012; Buckley et al., 2019).
Data from a national survey showed that about 90% of the general
population in Canada had detectable urinary BPA levels (>0.2 ng/mL)
(Health Canada, 2015), and in the US this percentage is even higher
(92.6%) (Calafat et al., 2008). BPA exposure is associated with a variety
of health problems, especially male reproductive disorders. Sing et al.
reported a reduction in the percentage of live sperm and a concomitant
increase in the percentage of dead sperm after exposure to different
doses of BPA (Singh et al., 2015). Analysis of testicular histology
revealed that in ICR mice pups postnatal BPA exposure led to a block in
the progression of meiosis and an increase in germ cell apoptosis during
the rst wave of spermatogenesis (Xie et al., 2016). Moreover, paternal
BPA exposure in CD-1 mice affected spermatogenesis by decreasing the
number stage of VIII seminiferous epithelial cells, causing a decline in
total sperm counts and sperm motility in the F1 offspring (Rahman and
Pang, 2019).
In males, spermatogenesis is a highly complicated process involving
mitosis of spermatogonia, meiosis of spermatocytes, and the differenti-
ation of round spermatids into mature sperm (Liu et al., 2021a). The
main regulators of spermatogenesis are the gonadotropic hormones
luteinizing hormone (LH) and follicle-stimulating hormone (FSH) which
are released by the pituitary in response to the hypothalamic derived
releasing neurohormone gonadotrophin-releasing hormone (GnRH)
(Corradi et al., 2016; Qiu et al., 2018). Furthermore, testosterone syn-
thesis in Leydig cells was mainly controlled by luteinizing hormone
(LH), which together with FSH regulates Sertoli cell function and the
normal progression of spermatogenesis within the seminiferous tubules
(Spaziani et al., 2021). Kisspeptin neurons in the hypothalamus act to
regulate the activity of GnRH neurons and thus, in turn, the downstream
hypothalamic-pituitary-gonadal (HPG) endocrine axis (Abbara et al.,
2021). The GnRH neurons in the hypothalamus are however not directly
able to respond to uctuations in circulating testosterone levels but
respond to changes in kisspeptin release, a neurotransmitter that binds
to the GPR54 receptor on the GnRH neurons. Kisspeptin is produced by
the kisspeptin neurons in the hypothalamus which express androgen
receptors and thus can respond via a negative feedback mechanism, to
changes in circulating testosterone levels (Poling and Kauffman, 2013).
BPA was able to bind androgen receptors (AR) and acted as an androgen
antagonist to block the effect of endogenous androgens, thereby
affecting the transmission of hormone signals in target cells, tissues, and
organs, resulting in the corresponding biological effects, and then
causing dysfunction of the reproductive system of the body (Murata and
Kang, 2018).
In addition, the histone-to-protamine exchange is the rst step dur-
ing post-meiotic male germ cell development, when haploid round
spermatids elongate and transform into spermatozoa. The failure of
histone-to-protamine exchange may contribute to male sterility (Gou
et al., 2017). The mammalian target of rapamycin (mTOR), belonging to
the phosphatidylinositol kinase-related kinase family, is a
serine-threonine protein kinase that is regulated by proline (Xu et al.,
2016). Activation of the phosphoinositide 3-kinase/protein kinase
B/mTOR (PI3K/AKT/mTOR) signaling pathway initiates the efcient
translation of mRNAs required for spermatogonial differentiation. At
present, studies investigating the impact of this pathway on testicular
function are numerous (Gao et al., 2020; Kong et al., 2021; Ni et al.,
2021; Fu et al., 2020).
The intestinal microbiota is a complex ecosystem in humans and
animals, and it has been found to play an important role in controlling
host health. Modulation of the intricate relationship between the host
and microbiome can result in many diseases such as cancer, metabolic,
cardiovascular, immune, and neurobehavioral disorders (Diamante
et al., 2021). Some exogenous compounds can disrupt gut microbiota
composition and induce negative health effects (Zhan et al., 2020).
Low-dose exposures to diethyl phthalate, methylparaben, and triclosan
decreased the body weight of adolescent rats, which coincides with a
decrease in the Firmicutes/Bacteroidetes ratio (Hu et al., 2016). More-
over, BPA is likely to inuence organisms via changes in microbial
companions. A previous study showed that exposure to BPA increased
the abundance of Lactobacilllus, Alcaligenes, and Mycobacterium in the
colon (Wang et al., 2018). Dietary exposure to BPA similarly altered the
gut microbiota composition as the intake of a diet high in fat and sucrose
does (Lai et al., 2016). In addition, BPA and ethinyl estradiol (EE) at
doses that have previously been shown to disturb behavior and meta-
bolism in F1 California mice offspring also led to generational and
sex-dependent changes in the gut microbiome (Javurek et al., 2016).
Despite the growing number of researches, the underlying mecha-
nisms between BPA-induced reproductive toxicity and gut microbiota
are still limited. Therefore, this study aimed to explore the potential link
between BPA-induced intestinal ora disturbance and reproductive
toxicity, further provided new insight into the comprehensive under-
standing of the mechanism of reproductive toxicity of BPA.
2. Materials and methods
2.1. Animals and experimental design
Male Sprague-Dawley rats (34weeks) were obtained from Beijing
HFK Bioscience Co., LTD. The animals were housed under temperature
(2225 C) and humidity (40%60%) controlled conditions with a 12 h
light/dark cycle. Rats had ad libitum access to chow and water. BPA
(analytical purity99%, CAS: 80057) was purchased from Sigma-
Aldrich (St. Louis, Missouri, US). After a 7-day adaptation period, the
rats were randomly divided into four groups (8 rats/group) and treated
as follows: control group (corn oil only), BPA-low (BPA-L), -medium
(-M), and -high (-H) group (30, 90, and 270 mg/kgbw BPA dissolved in
corn oil, respectively).
The no observed adverse effect level (NOAEL) and the lowest
observable adverse effect level (LOAEL) of BPA were 5 mg/kgbw and
50 mg/kgbw, respectively. Here, we chose the low doseof BPA 50
mg/kgbw, and the medium and high-doseof BPA is the using con-
centrations >50 mg/kg bw referring to previous literature (Adegoke
et al., 2022; Peretz et al., 2014). BPA was provided to the rats by daily
oral gavage for 30 days. The animal experiment was approved by the
Animal Care and Protection Committee of Jinan University (Approval
No. IACUC-2020102712) and was performed by the US NIH Guide for
the Care and Use of Laboratory Animals (No. 8023, revised in 1978).
At the end of the experiment, all animals were anesthetized by
pentobarbital and sacriced. Blood was collected from the abdominal
aorta for further analysis. The testis and epididymis were quickly
removed and weighed. The left testis was snap-frozen and stored at
80 C until further analysis. The right testis and epididymis were xed in
modied Davidsons uid (Scientic Phygene) and 4% para-
formaldehyde respectively for further histological analysis. The hypo-
thalamus and pituitary were collected, frozen on dry ice, and stored at
80 C for RT-qPCR analysis. The testis and epididymis coefcients were
calculated in the following way:
testis coefficient =testis weight(g)
body weight(g)×100%
R. Liu et al.
Ecotoxicology and Environmental Safety 239 (2022) 113623
3
epididymis coefficient =epididymis weight(g)
body weight(g)×100%
2.2. Sperm analysis
The left cauda epididymis was used for the evaluation of sperm pa-
rameters. The cauda epididymis was trimmed of residual adipose tissue,
and then put into a culture dish with 10 mL preheated normal saline
(37 C), cut into small pieces to release the stored sperm, and kept at a
temperature of 37 C for 2 min. After that, 10
μ
L sperm suspension was
subjected to analyze the sperm count, and sperm morphology analysis
using a computer-aided semen analysis system (CASA, mL-608ZII, Song
Jing Tian Lun Biotechnology Co., Ltd.). At least 30 different elds were
analyzed for each sample.
2.3. Determination of oxidative stress in serum and testis
To determine oxidative stress in testis tissue, 0.1 g testis tissue, 10%
(w/v) was homogenized in PBS buffer (pH 7.4). The homogenate was
centrifuged at 1520g for 20 min after which the supernatant was
collected. The assay kits for the detection of superoxide dismutase
(SOD), glutathione (GSH), catalase (CAT), and malondialdehyde (MDA)
were purchased from Nanjing Jiancheng Bioengineering Institute. The
protein concentration of the testis samples was determined by bicin-
choninic acid (BCA) protein assay kit (Applygen Technologies Inc.,
China).
2.4. Histological and immunohistochemical analysis of testis and
epididymis
The left testes and epididymides were xed for 24 h at room tem-
perature, dehydrated in graded ethanol and xylene, and embedded in
parafn. The embedded tissues were cut into 4 µm thick sections and
stained with hematoxylin-eosin (H&E).
Immunohistochemical analysis of mTOR was performed as described
previously (Jiang et al., 2018). In brief, tissue sections were deparaf-
nized after which antigen retrieval in citrate buffer (pH 6.0) was per-
formed. The next sections were blocked with 3% hydrogen peroxide in
methanol for 15 min at room temperature to block endogenous perox-
idase activity and washed three times in phosphate-buffered saline (PBS)
(pH 7.4). After blocking the nonspecic sites with 3% BSA, PI3K (1:200,
Cat# 205841-AP, Proteintech), p-AKT (1:250, Cat# ab81283, Abcam),
and mTOR (1:1000, Cat# 668881-Ig, Proteintech) primary antibodies
were applied to the separate sections, and slides were kept overnight at
4C respectively. Subsequently, sections were incubated with the sec-
ondary antibody for 50 min, followed by DAB staining, and counter-
staining with hematoxylin for 3 min mTOR labeling was examined by a
microscope (NIKON ECLIPSE E100, Japan) equipped with an imaging
system (NIKON DS-U3). To evaluate the amount of cell staining, the
images were analyzed using an image analysis software (Image Pro-Plus
6.0, Media Cybernetics, MD, U.S.A.).
2.5. Determination of hormone levels
Dihydrotestosterone (DHT), FSH, LH, estradiol (E2), inhibin B
(INHB), testosterone, and testicular fructose levels were measured in
serum, while GnRH content was measured in hypothalamic tissue ho-
mogenates, using enzyme-linked immunosorbent assay (ELISA) kits. The
ELISA kits for GnRH, DHT, FSH, LH, E2, and INHB were purchased from
Jiang Lai Biotechnology Co., Ltd. (Shanghai, China). The ELISA kits for
testosterone and fructose were purchased from Cusabio (Wuhan, China)
and Nanjing Jiancheng Bioengineering Institute (Nanjing, China),
respectively. All procedures were carried out according to the manu-
facturersinstructions.
2.6. Determination of apoptosis rate in testes
Apoptosis of cells in testicular sections was detected using a terminal-
deoxynucleotidyl transferase-mediated nick end labeling (TUNEL)
staining following the manufacturers protocols of the assay kit from
Beyotime Biotech (Shanghai, China).
2.7. Quantication of mRNA expression
Total RNA from the hypothalamus and testis was extracted using
TRIzol reagent (Beyotime Institute of Biotechnology, China) following
the manufacturers protocol. The cDNA used for quantitative RT-PCR
was synthesized using the Evo M-MLV RT Kit with gDNA Clean for
qPCR II AG11711 (Accurate Biotechnology [Hunan] Co., Ltd, China).
RT-PCR reactions were carried out employing the Applied Biosystems
QuantStudio 6 using the SYBR® Green Premix Pro Taq HS qPCR Kit
AG11701 (Accurate Biotechnology [Hunan] Co., Ltd, China) according
to the following procedure: initial temperature of 95 C for 30 s, fol-
lowed by 5 s at 95 C (45 cycles) for denaturation and annealing at 55 C
for 30 s. The primers were synthesized by Shanghai Sangon Biological
Engineering Co., Ltd (Guangzhou synthesis department). Relative
mRNA expression levels were calculated using the 2
-ΔΔCT
method and
were normalized with β-actin. The primers sequences used are listed in
Table 1.
2.8. Protein expression analysis
Protein extraction and western blot analysis were performed as
previously described (Li et al., 2020). In brief, the testis tissues were
homogenized in RIPA buffer (Beyotime, China) supplemented with
protease inhibitors and phenylmethanesulfonyl uoride (PMSF), fol-
lowed by centrifugation at 14,000g for 10 min at 4 C after which the
supernatant was collected. The BCA protein assay kit was used to
determine the protein content of the samples. Equal amounts of protein
(20
μ
g) were separated by 812% sodium dodecyl
sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and transferred
onto polyvinylidene diuoride (PVDF) membranes. Membranes were
blocked with 5% skimmed milk at room temperature for 1.5 h and
incubated with the primary antibodies overnight at 4 C. The primary
antibodies Bax (1:500, Cat# AF1020), Bcl-2 (1:500, Cat# AF6139),
eIF4E (1:500, Cat# AF6110), FAS (1:500, Cat# AF5342), FASL (1:500,
Cat# AF0157), Histone H4 (1:500, Cat# AF6355), and β-actin (1:5000,
Cat# T0022) were acquired from Afnity Biosciences (USA). Histone
H2A (1:5000, Cat# ab177308), Histone H2B (1:1000, Cat# ab52484),
Histone H3 (1:1000, Cat# ab176882), and phospho-AKT (1:5000, Cat#
ab81283) was purchased from Abcam (Cambridge, UK). Cleaved-PARP
(1:1000, Cat# 94885 S), PARP (1:1000, Cat# 9532 T), ub-H2A (1:2000,
Cat# 8240 T), ub-H2B (1:1000, Cat# 5546 T), and were obtained from
Cell Signaling Technology (Massachusetts, USA). mTOR (1:5000, Cat#
668881-Ig), Raptor (1:500, Cat# 209841-AP), AKT (1:1000, Cat#
101762-AP), and PI3K (1:500, Cat# 205841-AP) were purchased from
Table 1
Primers used for RT-PCR.
Genes Sequences (5-3) Accession No.
Kiss-1 Forward: TGCTGCTTCTCCTCTGTGTG NM_181692.1
Reverse: CCAGGCATTAACGAGTTCCT
Gpr54 Forward: GGAACTCACTGGTCATCTTCGT NM_023992.2
Reverse: GTACGCAGCACAGAAGGAAAGT
P13k Forward: GATGTCTGCGTTAGGGCTTACC NM_001371300.2
Reverse: TCAGCATCATGGAGAACAGGAT
Akt1 Forward: CTCATTCCAGACCCACGAC NM_033230.3
Reverse: ACAGCCCGAAGTCCGTTA
mtor Forward: AGCCGTTGTTGCAGAGACTT NM_019906.2
Reverse: CATGGTTCATGGTGTCTTGC
β-actin Forward: CCCATCTATGAGGGTTACGC NM_031144.3
Reverse: TTTAATGTCACGCACGATTTC
R. Liu et al.
Ecotoxicology and Environmental Safety 239 (2022) 113623
4
Proteintech (Wuhan, China). Then it was followed by incubation with
goat anti-rabbit IgG-HRP (1:2000, Cat# 98164 S) (Cell Signaling Tech-
nology, USA) or anti-mouse IgG-HRP (1:5000, Cat# 91196 S) (Pro-
teintech, Wuhan, China) at room temperature for 1 h, as described
previously (Jiang et al., 2018). ECL reagent was applied to visualize the
bands by the Clinx ChemiScope system (Shanghai, China).
2.9. Gut microbiota analysis by 16S rRNA gene sequencing
Fecal samples were after collection stored at 80 C until further
processing. Total DNA was extracted from the fecal samples using the
MagPure Soil DNA LQ Kit (Magen, China) according to the manufac-
turers instructions. DNA concentration and quality were assessed by
NanoDrop 2000 spectrophotometry (Thermo Fisher Scientic, USA) and
agarose gel electrophoresis, respectively. For bacterial diversity anal-
ysis, the V3-V4 variable region of the 16 S rRNA genes was amplied
using the universal primers 338 F (5-ACTCCTACGGGAGGCAGCA-3)
and 806 R (5-GGACTACHVGGGTWTCTAAT-3). Amplicon quality was
visualized using gel electrophoresis. The PCR products were puried
using Agencourt AMPure XP beads (Beckman Coulter Co., USA) and
quantied using the Qubit dsDNA assay kit (Life Technologies, USA).
Puried amplicons were pooled in equimolar concentrations.
Sequencing was performed on the Illumina NovaSeq 6000 sequencing
platform with the paired-end read of 2 ×250 cycles according to stan-
dard protocols (Illumina Inc., San Diego, CA; OE Biotech Company;
Shanghai, China).
For bioinformatic analysis paired-end reads were assembled using
FLASH (FLASH: fast length adjustment of short reads to improve genome
assemblies). Further processing of paired-end reads including quality
ltering, removal of mismatched barcodes, and sequences were
completed using QIIME version 1.8.0. Clean reads were subjected to
primer sequence removal and clustering to generate operational taxo-
nomic units (OTUs) using Vsearch software with a 97% similarity cutoff.
All representative reads were annotated and blasted against Silva
database Version 132.
2.10. Statistical analysis
Statistical analyses were conducted using SPSS 23.0 (SPSS Inc.,
Chicago, USA). GraphPad Prism software, version 8.0 (San Diego, USA)
and Origin, version 9.1 (OriginLab Corp., Northampton, MA) were used
for gure design. A signicant difference was determined by one-way
Fig. 1. Effects of BPA exposure on sperm quality, and oxidative stress in SD rats. (A) Body weight change. (B) Testis weight. (C) Epididymis weight. (D) Testis
coefcient. (E) Epididymis coefcient. Sperm quality and morphology were analyzed by CASA. (F) Sperm counts. (G) Sperm abnormality. (H) Representative ex-
amples of sperm morphology. Abnormalities characterized by deformed or absent head (black arrows) and docked tail (black triangle) are seen in rats. (I) The levels
of superoxide dismutase activity (SOD), glutathione (GSH), catalase activity (CAT), and malondialdehyde (MDA) in rat serum. (J) The levels of SOD, GSH, CAT, and
MDA in rat testes. Data are expressed as mean ±SD (n =8). Bars with different letters represent a signicant difference between groups (P<0.05).
R. Liu et al.
Ecotoxicology and Environmental Safety 239 (2022) 113623
5
analysis of variance (ANOVA), and the differences between the two
groups were analyzed by the Bonferroni post hoc test. Values were
considered to be signicantly different when P<0.05.
3. Results
3.1. Effects of BPA exposure on anthropomorphic parameters and sperm
quality
Exposure to BPA led to a lower increase in the body weight of the rats
over time when compared to the control animals (Fig. 1A). Body, testis,
and epididymis weights and the coefcients of testes and epididymides
were however not signicantly affected by BPA treatment (Fig. 1B-E).
Exposure to the high dose of BPA (BPA-H) notably decreased total sperm
count compared with the controls (P<0.05) (Fig. 1F, and H).
Concomitantly, the sperm malformation ratio was signicantly
increased in the BPA-M, and BPA-H groups, characterized by the
headless and tail-docking sperm (P<0.05) (Fig. 1G).
3.2. Effects of BPA exposure on serum and testicular oxidative stress
markers
To elucidate the presence of oxidative stress induced by BPA expo-
sure, the levels of MDA and antioxidant markers SOD, GSH, and CAT
were measured in rat serum and testis tissue homogenates. A signicant
decrease in serum SOD and CAT levels was observed in rats of the BPA-H
group compared to the controls (P<0.05) (Fig. 1I). GSH level was
signicantly decreased in the BPA-M group (P<0.05) (Fig. 1I), while
MDA content in serum and testis was higher in the BPA-M and BPA-H
groups when compared to the control animals (P<0.05) (Fig. 1I, J).
As shown in Fig. 1J, exposure to BPA had no signicant effects on the
testicular levels of SOD and CAT, while a signicant decrease in GSH
level was observed in testis tissue in the BPA-M and BPA-H groups.
3.3. Effects of BPA exposure on testis and epididymis histology
As shown in Fig. 2A, the testis tissue showed a regular arrangement
of different stages of germ cells in the seminiferous tubules of the control
group in rats. Sertoli cells and spermatogonia were located on intact
basement membranes. The slides from BPA-treated groups showed
disorganization of the germinal epithelial layers of the seminiferous
tubules. In the BPA-exposed groups, some seminiferous tubules showed
mild atrophy as evidenced by the presence of vacuoles between the germ
cells, while the interstitial space seemed enlarged. Maturation of sperm
largely occurs in the epididymis; in the cauda epididymis, the sperm
acquires motility. Fig. 2A gives the impression that sperm numbers are
reduced in the BPA-M and BPA-H treated animals when compared to the
controls, which is conrmed for the BPA-H group by the total sperm
count shown in Fig. 1F.
3.4. Effects of BPA exposure on hormone levels
In this study, medium and high dosages of BPA administration
signicantly upregulated hypothalamic Kiss1 mRNA expression
(P<0.05) (Fig. 2B). However, Fig. 2C revealed that there was a sig-
nicant decrease in GPR54 mRNA expression in low and high groups.
GnRH protein content in the hypothalamus was also signicantly higher
in the BPA-treated rats compared to the control group (Fig. 2D). Serum
testosterone levels were reduced in the BPA-M and BPA-H groups
compared to the untreated controls (P<0.05) (Fig. 2E). Similarly,
serum INHB level was signicantly decreased in the BPA-H group
(P<0.05) (Fig. 2I), while FSH and DHT levels were signicantly
Fig. 2. Effects of BPA exposure on histology of testis and epididymis, and hormone levels. (A) Testis and epididymis. From left to right: control, BPA-L, BPA-M, BPA-
H. Bar represents 50 µm and 100 µm, respectively. RT-PCR analyses of the mRNA levels of (B) Kiss-1, and (C) Gpr54 in the hypothalamus. (D) GnRH protein content in
the hypothalamus. The levels of (E) testosterone, (F) E2, (G) luteinizing hormone (LH), (H) follicle-stimulating hormone (FSH), (I) inhibin B (INHB), and (J)
dihydrotestosterone (DHT) in rat serum was measured by ELISA. Data are expressed as mean ±SD (n =8). Different letters represent a signicant difference between
groups (P<0.05).
R. Liu et al.
Ecotoxicology and Environmental Safety 239 (2022) 113623
6
elevated in the BPA-M and BPA-H groups (P<0.05) (Fig. 2H, J). There
was no signicant difference in E2 and LH levels among the different
groups.
3.5. Effects of BPA exposure on PI3K/AKT/mTOR pathway in testis
To determine whether chronic BPA administration affects the PI3K/
AKT/mTOR signaling pathway and in this way affects testis function, we
investigated the relative expressions of key proteins and genes in this
pathway by Western blotting and RT-PCR. As shown in Fig. 3A-C,
exposure to BPA had no signicant effect on the gene expression of Pi3k,
Akt, and mtor. However, the protein levels of the PI3K in the high-dose
group were signicantly escalated in testis (P<0.05) (Fig. 3D), as well
as the level of p-AKT in medium and high dosage groups (P<0.05)
(Fig. 3F). Accordingly, the expression of PI3K and p-AKT determined by
immunohistochemically staining were upregulated in BPA-treated
groups. Moreover, we evaluated the localization of mTOR in the testis
by immunohistochemistry. mTOR expression in rat testicular tissue was
seen in Sertoli cells, spermatogonia, spermatocytes, and some Leydig
cells. The protein level of mTOR was signicantly increased in the
testicular after being treated with medium and high dosage BPA
compared with the control group (Fig. 3L). The protein expression of the
Raptor was increased in the BPA-H group compared to the control group
(P<0.01) (Fig. 3I). Interestingly, the protein expression of 4EBP1 was
signicantly reduced in the BPA-M and BPA-H groups compared to the
group (P<0.05) (Fig. 3J), while the protein level of eIF4E was signi-
cantly increased (Fig. 3K).
3.6. Effects of BPA exposure on apoptosis in testes
Apoptosis is one of the main forms of cell death in which mito-
chondria play a key role. To explore whether chronic BPA exposure
leads to an increase in testicular apoptosis, we examined the expression
of apoptosis-related proteins by western blot. As illustrated in Fig. 4A-G,
a signicant increase was observed in cleaved-PARP/PARP, FAS, and
Caspase3 levels after exposure to the high dosage of BPA. No signicant
difference was observed in the levels of FASL, Bax, and Bcl-2 compared
to the control group (P<0.05) (Fig. 4C, F, and G). In addition, apoptotic
cells in the testes section were subsequently detected by TUNEL staining.
As shown in Fig. 4H, BPA-induced apoptosis occurs in both Leydig cells
and germ cells which is characterized by spermatogonia and sper-
matocytes compared to the control. These results indicated that BPA
treatment may increase the incidence of apoptosis to some extent.
3.7. Effects of BPA exposure on histone ubiquitination in testes
To comprehend the expression of H2A, H2B, ub-H2A, ub-H2B, H3,
and H4 in the testes, western blotting was applied to observe the ex-
pressions of these parameters in rats from different groups. H2A and H3
expressions were higher in the medium and high groups (P<0.05,
P<0.01) (Fig. 5A, B, and F), while ubiquitin histone H2A (ub-H2A) and
ubiquitin histone H2B (ub-H2B) was markedly reduced in BPA-exposure
groups compared with the control group (Fig. 5A, D, and E). There was
no signicance in the H2B and H4 protein expressions (Fig. 5C, G).
3.8. Effects of BPA exposure on gut microbiota composition
To determine whether BPA treatment affects gut microbiota
composition, we analyzed the 16S rRNA gene sequence of the micro-
biota. Generally,
α
-diversity reects the abundance of the gut micro-
biota, and β-diversity is used to evaluate the community similarity and
diversity of different groups. As shown in Fig. 6A, the PCA reecting the
β-diversity showed an obvious separation among the groups, which
means that the intestinal microbiota composition changed signicantly
after treatment with BPA. The BPA-M and BPA-H groups exhibited
signicantly lower Chao1 and higher goods coverage (P<0.05,
P<0.01) (Fig. 6B, C). Shannon (Fig. 6D) showed a downward trend in
medium and high groups, implying that BPA exposure may lead to lower
intestinal microbiota diversity. The composition of gut microbiota
among the groups at the phylum, class, order, and family levels is shown
in Fig. 6E-H. At the phylum level, the main gut microbiota were Bac-
teroidetes (66.21% vs 73.96% vs 71.71% vs 63.3%), Firmicutes (26.23%
vs 22.65% vs 23.26% vs 29.06%), Proteobacteria (2.48% vs 2.26%
vs2.63% vs 4.90%), and Epsilonbacteraeota (4.47% vs 0.5% vs 1.71% vs
1.85%) in Control, BPA-L, M, and H groups, respectively. The relative
abundance of class Campylobacteria were 4.45%, 0.6%, 1.69%, and
1.80%, whereas Gammaproteobacteria were 0.83%, 1.23%, 1.81%, and
3.97% in Control, BPA-L, M, and H groups, respectively (Fig. 6F). At the
order level, the BPA-L group showed a lower abundance of Campylo-
bacterales (0.48%) compared to the control rats (4.37%). In addition, the
relative abundance of Betaproteobacteriales was increased in the BPA-H
group (2.32%) compared to the control rats (0.63%) (Fig. 6G). How-
ever, there was no signicant difference in the main microbiota at the
family level among groups.
3.9. Correlations between the intestinal microbiota and hormone
parameters
To conrm the signicant differences in microbiota composition and
identify biomarkers in intestinal microbiota among the treatment
groups, we applied LEfSe, an algorithm for microbial marker detection.
As depicted in Fig. 7A and B, the differential enrichment of specic
bacteria was shown in both cladograms and histograms based on an LDA
score >3. For the control group, the genus Prevotellaceae_Ga6A1_group,
and genus Lachnospiraceae_NK4A136_group were the dominant micro-
biota, while the genus Alloprevotella, genus uncultured_organism, and
genus Prevotellaceae_UCG_001 appeared to be dominant in the BPA-M
group. The LEfSe histogram showed further that the BPA-H group con-
tained the largest number of bacterial taxa, suggesting that the BPA
effect on the bacterial community composition mainly depended on the
doses. Furthermore, we observed that the relative abundance of the class
Gammaproteobacteria and the genus Parasutterella, family Burkholder-
iaceae, order Enterobacteriales were signicantly higher in the BPA-H
group than in the control group, suggesting that these may serve as
taxonomic biomarkers.
The correlation analysis with a heatmap indicated that the relative
abundance of several key intestinal microbial phylotypes was signi-
cantly correlated with the reproductive hormones.
Parameters (Fig. 7C). Fusicatenibacter was notably positively corre-
lated with serum DHT, E2 levels, and kiss-1 expression in the hypo-
thalamus, while a negative correlation was observed with testosterone
levels. Desulfovibrio, Ruminiclostridium, Acetatifactor, Rikenella, and
Ruminiclostridium_5 were negatively correlated with serum FSH, LH,
DHT, and E2 levels, and kiss-1 expression in the hypothalamus, and
positively correlated with serum INHB and testosterone levels, GnRH
protein level and Gpr54 gene expression in the hypothalamus. Overall,
most of the bacterial genera of composition were altered by BPA
administration and exhibited strong correlations with hormones.
4. Discussion
Reports have demonstrated that BPA is a part of many aspects of
daily living, as a variety of common consumer goods ranging from water
bottles, dinner plates, and CDs (Wang et al., 2014). BPA can disrupt the
endocrine function related to hormone levels due to its estrogenic and
antiandrogenic activity and is known to affect male fertility (Cariati
et al., 2019). Although the effect of BPA on the testis has been studied,
whether BPA-induced male reproductive toxicity is associated with the
alteration of gut microbiota has remained largely unknown. In the
present study, in vivo experiment with multiple dimensions analysis was
conducted to elucidate the potential mechanism of BPA exposure on the
reproductive system in male rats.
R. Liu et al.
Ecotoxicology and Environmental Safety 239 (2022) 113623
7
Fig. 3. Effects of BPA exposure on PI3K/AKT/mTOR pathway in testis (A-C) The mRNA expression levels of Pi3k, Akt1, and mtor in testis. (D-K) The protein
expression levels of PI3K, mTOR, p-AKT, p-AKT/AKT, Raptor, 4EBP1, and eIF4E in testis. Different letters indicate signicant differences between groups (P<0.05).
(L) Immunohistochemical staining for PI3K, p-AKT, and mTOR proteins in testes tissue (200×, 400×magnications). Quantitative data are presented as mean ±SD
(n =4). * P<0.05, ** P<0.01, compared with control group.
R. Liu et al.
Ecotoxicology and Environmental Safety 239 (2022) 113623
8
Fig. 4. Effects of BPA exposure on cell apoptosis in testes. (A) Western blotting photographs and relative quantitative analysis of (B) Cleaved-PARP and PARP
protein, (C) FASL, (D) FAS, (E) Cleaved-caspase3, (F) Caspase3, (G) Bcl-2, (H) Bax, and (I) Bcl-2/Bax. Data are expressed as mean ±SD (n =4). Different letters
indicate signicant differences between groups (P<0.05). (H) The assessment of apoptosis in testes sections with TUNEL immunouorescence and the ratio of
apoptosis cells were counted in every eld. Quantitative data are presented as mean ±SD (n =4). * P<0.05, ** P<0.01, compared with control group.
Fig. 5. Effects of BPA exposure on the levels of histone ubiquitination in testes. (A) Representative western blots and relative quantitative analysis of (B) H2A, (C)
H2B, (D) ub-H2A, (E) ub-H2B, (F) H3, and (G) H4. The data are expressed as mean ±SD (n =4). Different letters indicate signicant differences between
groups (P<0.05).
R. Liu et al.
Ecotoxicology and Environmental Safety 239 (2022) 113623
9
Spermatogenesis is a highly ordered and precisely regulated process,
which requires huge epigenetic remodeling (Govin et al., 2006). Normal
spermatogenesis is dependent on well-balanced spermatogenetic cell
proliferation, differentiation, and death in the testes (Zhang et al.,
2012). LH stimulated the production of testosterone by Leydig cells, and
FSH stimulated the local production of estradiol by Sertoli cells. Besides,
FSH and testosterone directly control the expression of genes in Sertoli
cells which are required for the progression of meiotic and post-meiotic
events (OShaughnessy et al., 2010). Normal testosterone levels are
necessary for maintaining the development of the male reproductive
system (Zhao et al., 2020). In our study, a high dose of BPA-treated rats
showed less sperm count while a higher deformity rate signicantly.
And the decreased levels of testosterone further conrmed the disrup-
tion of hormones involved in the BPA-induced testicular toxicity. The
testicular normal structure is important for maintaining normal
reproductive function. And the histopathological examination of testis
and epididymis is also important for fertility diagnosis and prognosis
inference in reproductive practice (McLachlan et al., 2007). Consistent
with the data on sperm quality, we observed disarrangement of testic-
ular tissue, and atrophy of spermatogenic tubules in a dose-dependent
manner, suggesting that BPA inuences spermatogenesis.
The kisspeptin system is a key regulator of the reproductive system,
and the formation and function of the kisspeptin signaling pathway are
affected by sex hormones at different stages of the life cycle, which can
regulate the reproductive endocrine system. Kiss-1 gene, as a target for
regulation by testosterone, the increased expression of GnRH, and FSH
can also be understood as negative feedback regulation of testosterone.
The upregulation of hypothalamic Kiss-1 after BPA exposure may
stimulate the synthesis and release of GnRH and gonadotropins in the
hypothalamus and pituitary gland, respectively (Xi et al., 2011). In the
Fig. 6. Effects of BPA exposure on gut microbiota composition of rats. (A) Principal component analysis (PCA) of gut bacterial community composition.
α
-diversity
analysis: (B) Chao1 index, (C) Shannon index, and (D) Simpson index. Composition of microbial community and the main microbiota at (E) phylum, (F) class, (G)
order, and (H) family level. Statistical signicance was determined by one-way ANOVA and the LSD post-hoc test, * P<0.05, ** P<0.01 compared to the con-
trol group.
R. Liu et al.
Ecotoxicology and Environmental Safety 239 (2022) 113623
10
present study, we observed that BPA exposure decreased INHB levels in
serum, and increased the FSH and DHT levels. The decrease in INHB may
be indicative of a BPA-induced alteration of Sertoli cell functionality.
The effects on seminiferous epithelium morphology are relatively mild
which may be due to a compensatory effect by DHT, a metabolite of
testosterone that binds with a substantially higher afnity to the
androgen receptor than testosterone (Carson and Rittmaster, 2003).
Moreover, in line with the increased Kiss-1 expression in the hypotha-
lamic, GnRH gene expression in the pituitary increased, which is similar
to a study by Oride et al. who showed that clomiphene citrate induced
Kiss-1 expression in the presence of estradiol in mHypoA-50 cells (Oride
et al., 2020). These results suggest that BPA exposure disrupted the
imbalance of hormones, further leading to a suboptimal maturation of
the spermatids and being responsible for the observed increased sperm
malformation.
As another concerned item, the process of spermatogenesis was
regulated by histone modication accurately, and the histone ubiquiti-
nation process is the master prerequisite of nucleosome removal during
the sperm elongating period (Zhao et al., 2022). As the rst step in
histone-to-protamine exchange, histone ubiquitination can promote
histone removal by loosening the compact nucleosome, subsequently,
promoting the replacement of transition protein in pachytene sper-
matocyte (Li et al., 2020). In our study, the protein expression of H2A
and H3 were signicantly increased in medium and high doses of
BPA-treated groups, while the ubiquitinated histone H2A and ubiquiti-
nated H2B were signicantly diminished. Therefore, these data indi-
cated BPA exposure affected spermatogenesis by altering the
ubiquitination.
Testicular cells contain a large amount of unsaturated fatty acids and
divide at a high speed, which makes spermatogenic cells highly sus-
ceptible to oxidative damage, which is not conducive to sperm pro-
duction (Sharma et al., 2019). Reactive oxygen species (ROS) are
involved in DNA damage, resulting in the status of oxidative stress and
possibly modulating the apoptotic pathway. SOD can catalyze the
overproduced O
2
into H
2
O
2
, next H
2
O
2
is catalyzed into H
2
O and O
2
by
CAT, which protected the body from harmful substances-induced
oxidative stress. GSH is a crucial non-protein antioxidant and can
scavenge the lipid peroxide radicals. When an exogenous substance
enters the body, it will be slowly released into the blood, and then reach
organs through the blood. Therefore, the oxidation indicators in the
blood are the rst to be observed when the body is oxidized and injured.
That is the reason that the SOD and CAT activities were signicantly
increased in the blood compared to the control group in this study.
While the lipid peroxidative products exceed the scavenging efciency
of GSH, it will accumulate in the blood and tissues, and there is a sig-
nicant increase in GSH. Histones can migrate from the nucleus into the
cytoplasm as a response to DNA double-strand breakage and then can
indirectly activate BAK at the mitochondrial outer membrane, resulting
in the promotion of cytochrome C release and activation of a cell pro-
grammed apoptosis. In our study, we found that the protein levels of
Cleaved-PARP/PARP and FAS were signicantly increased in the BPA-H
group, as well as the percentage of TUNEL-positive cells in rat testes
expoto sed BPA. Cleaved-caspase3, which is the main nal executor of
apoptosis, is responsible for the cleavage of key cellular proteins, leading
Fig. 7. Correlations between the intestinal microbiota and hormone parameters. (A) Cladograms based on LEfSe analysis representing featuring bacterial taxa. (B)
LDA score, each taxon with a threshold score larger than 3 is shown in the histogram, the bar length of LDA represents the impact of signicantly different species in
each group. (C) Correlation analysis between intestinal ora and hormones in rats. Spearmans correlation coefcients are represented by color ranging from blue,
negative correlation (0.5), to red, positive correlation (0.5). Signicant correlations are noted by * P<0.05, ** P<0.01, and *** P<0.001.
R. Liu et al.
Ecotoxicology and Environmental Safety 239 (2022) 113623
11
to apoptosis (Budihardjoetal.,1999). Increased activities of
Cleaved-caspase3 in this study proved that caspase cascades are
involved in BPA-induced spermatogenic cell apoptosis. However, there
was no signicant change in Bcl-2 and Bax. We speculated that the
apoptosis of testis tissue induced by BPA may be mainly affecting the
FAS pathway rather than the Bax pathway.
The process of germ cell development is under the tight control of
various signaling pathways, including the PI3K/Akt/mTOR pathway. A
previous study has conrmed that PI3K/AKT/mTOR signaling pathway
is one of the classical pathways to inhibit apoptosis (Wu et al., 2020).
Moreover, FSH can regulate Sertoli cell proliferation through the
pathway (Riera et al., 2012). The germ cells in the testis are highly
proliferative and metabolically active. It is therefore not surprising that
the PI3K/AKT/mTOR pathway plays a central role in the self-renewal of
spermatogonial stem cells and the proliferation and differentiation of
spermatogonia (Moreira et al., 2019; Cao et al., 2020; Zhang et al.,
2020). Therefore, we next evaluated the PI3K/AKT pathway. The cur-
rent data show that BPA exposure leads to activation of the PI3K/AKT
pathway in testes of male SD rats. Meanwhile, the inhibition of mTOR
may interfere with Sertoli cell functionality, leading to the premature
release of sperm cells (Boyer et al., 2016). Therefore, we speculate it is
probably a protective compensation reaction, that is the activation of
AKT can protect cells from damage under oxidative stress.
Gut microbiota plays a role in metabolic disorders and participates in
the regulation of hormonal levels, estrous cycle, and reproductive
functions (Hussain et al., 2021). A previous study showed that gut mi-
crobes can involve in estrogen and androgen cycling and inuence sex
steroid hormone levels, as well as androgen production from glucocor-
ticoids (Cross et al., 2018). Sexual maturation was a major determinant
of the cecal microbiome community structure, and microbial coloniza-
tion status was correlated with testosterone levels. The removal of the
gut microbiota decreases testosterone levels in male mice, indicative of
bidirectional interaction between the amount of male sex hormone and
the microbiota (Markle et al., 2013). In addition, a previous study
indicated that testosterone may be one of the indirect factors leading to
the development of amphoteric colon adenoma (Amos-Landgraf et al.,
2014). In turn, the gut microbiota itself also inuences estrogen levels
(Rizzetto et al., 2018). The present study showed that BPA exposure
caused gut microbial dysbiosis, with lower diversity in
α
-diversity and
altered relative abundance for certain bacterial taxa. Bacteroidetes, and
Firmicutes, which accounted for up to 90% of the total sequences, were
the dominant phylum in BPA-treated groups. The relative abundances of
the class Gammaproteobacteria and the order Betaproteobacteriales were
elevated after exposure to the high dose of BPA. Ni et al. (2021) have
found that BPA exposure increases the abundance of Firmicutes and de-
creases the abundance of Bacteroidetes in C57BL/6 male mice. Proteus is
anaerobic or facultative anaerobic bacillus in the gastrointestinal tract
and hydrolyzes urea into ammonia and carbon dioxide via urease (Fan
et al., 2020). Gammaproteobacteria are involved in the synthesis of
vitamin B12 and have multiple benecial functions in the intestine
including the synthesis and uptake of amino acids (Paris et al., 2020).
Therefore, we speculate that BPA may increase the permeability of the
intestinal barrier by altering the gut microbiome. This pathological
change may induce the likelihood that bacterial pathogens will enter the
systemic circulation, which may affect the secretion of hormones in the
body (Ait-Belgnaoui et al., 2012).
In future research, the contents of different bacteria and multi-omic
techniques such as proteomics, and metabolomics are needed to reveal
novel interactions between gut microbiota and sex hormones.
5. Conclusion
In summary, BPA-induced gut microora disorder is closely related
to male reproductive toxicity. Disorder of sex hormone levels is strongly
associated with the imbalance of intestinal ora. BPA exposure affected
spermatogenesis by increasing oxidative stress and leading to
mitochondria apoptosis. In addition, the histone modication was per-
turbed, and the PI3K/AKT pathway was activated in rat testicular after
exposure to BPA. Further research is needed to investigate the mecha-
nisms of those identied key microbiota as a biomarker for certain
reproduction-related diseases.
Funding
This work was supported by the Youth Science and Technology
Innovation Talent of Guangdong TeZhi Plan (No. 2019TQ05N770);
Guangdong Basic and Applied Basic Research Foundation (No.
2020A1515111045); Guangdong Key Area Research and Development
Program (No. 2019B020210003); Construction Plan of Guangdong
Province High-level Universities and the Research Start-up Funds for the
High-level Talent Introduction Project of South China Agricultural
University (No. 20173326).
CRediT authorship contribution statement
Ruijing Liu: Investigation, Formal analysis, Methodology, Writing
original draft. Dongbao Cai: Methodology, Data curation. Xusheng Li:
Data curation, Validation. Writing review & editing. Boping Liu:
Funding acquisition, Project administration, Writing review & editing.
Jiali Chen: Methodology. Xinwei Jiang: Methodology. Haiwei Li:
Methodology, Visualization. Zhenhua Li: Methodology. Katja Teerds:
Writing review & editing. Jianxia Sun: Data curation. Weibin Bai:
Conceptualization, Funding acquisition, Project administration, Re-
sources, Supervision. Yulong Jin: Data curation, Funding acquisition,
Writing review & editing, Supervision.
Declaration of Competing Interest
The authors declare that they have no known competing nancial
interests or personal relationships that could have appeared to inuence
the work reported in this paper.
References
Adegoke, E.O., Rahman, M.S., Amjad, S., Pang, W.K., Ryu, D.Y., Park, Y.J., Pang, M.G.,
2022. Bisphenol A damages testicular junctional proteins transgenerationally in
mice. Environ. Pollut. 302, 119067 https://doi.org/10.1016/j.envpol.2022.119067.
Ait-Belgnaoui, A., Durand, H., Cartier, C., Chaumaz, G., Eutamene, H., Ferrier, L.,
Houdeau, E., Fioramonti, J., Bueno, L., Theodorou, V., 2012. Prevention of gut
leakiness by a probiotic treatment leads to attenuated HPA response to an acute
psychological stress in rats. Psychoneuroendocrinology 37, 18851895. https://doi.
org/10.1016/j.psyneuen.2012.03.024.
Amos-Landgraf, J.M., Heijmans, J., Wielenga, M.C., Dunkin, E., Krentz, K.J., Clipson, L.,
Ederveen, A.G., Groothuis, P.G., Mosselman, S., Muncan, V., Hommes, D.W.,
Shedlovsky, A., Dove, W.F., van den Brink, G.R., 2014. Sex disparity in colonic
adenomagenesis involves promotion by male hormones, not protection by female
hormones. Proc. Natl. Acad. Sci. USA 111, 1651416519. https://doi.org/10.1073/
pnas.1323064111.
Boyer, A., Girard, M., Thimmanahalli, D.S., Levasseur, A., C´
eleste, C., Paquet, M.,
Duggavathi, R., Boerboom, D., 2016. mTOR regulates gap junction Alpha-1 protein
trafcking in sertoli cells and is required for the maintenance of spermatogenesis in
mice. Biol. Reprod. 95, 13. https://doi.org/10.1095/biolreprod.115.138016.
Buckley, J.P., Kim, H., Wong, E., Rebholz, C.M., 2019. Ultra-processed food consumption
and exposure to phthalates and bisphenols in the US National Health and Nutrition
Examination Survey, 2013-2014. Environ. Int. 131, 105057 https://doi.org/
10.1016/j.envint.2019.105057.
Calafat, A.M., Ye, X., Wong, L., Reidy, J.A., Needham, L.L., 2008. Exposure of the U.S.
population to bisphenol A and 4-tertiary-octylphenol: 2003-2004. Environ. Health
Perspect. 116, 3944. https://doi.org/10.1289/ehp.10753.
Cao, J., Lin, Z., Tong, M., Zhang, Y., Li, Y., Zhou, Y., 2020. Mechanistic target of
rapamycin kinase (Mtor) is required for spermatogonial proliferation and
differentiation in mice. Asian J. Androl. 22, 169176. https://doi.org/10.4103/aja.
aja_14_19.
Cariati, F., DUonno, N., Borrillo, F., Iervolino, S., Galdiero, G., Tomaiuolo, R., 2019.
Bisphenol a: an emerging threat to male fertility. Reprod. Biol. Endocrinol. 17, 6.
https://doi.org/10.1186/s12958-018-0447-6.
Carson, C., Rittmaster, R., 2003. The role of dihydrotestosterone in benign prostatic
hyperplasia. Urology 61, 27. https://doi.org/10.1016/s0090-4295(03)00045-1.
R. Liu et al.
Ecotoxicology and Environmental Safety 239 (2022) 113623
12
Cho, S., Choi, Y.S., Luu, H.M., Guo, J., 2012. Determination of total leachable bisphenol
A from polysulfone membranes based on multiple consecutive extractions. Talanta
101, 537540. https://doi.org/10.1016/j.talanta.2012.09.033.
Corradi, P.F., Corradi, R.B., Greene, L.W., 2016. Physiology of the hypothalamic
pituitary gonadal axis in the male. Urol. Clin. N. Am. 43, 151162. https://doi.org/
10.1016/j.ucl.2016.01.001.
Cross, T.L., Kasahara, K., Rey, F.E., 2018. Sexual dimorphism of cardiometabolic
dysfunction: gut microbiome in the play? Mol. Metab. 15, 7081.
Demierre, A., Peter, R., Oberli, A., Bourqui-Pittet, M., 2012. Dermal penetration of
bisphenol A in human skin contributes marginally to total exposure. Toxicol. Lett.
213, 305308. https://doi.org/10.1016/j.toxlet.2012.07.001.
Diamante, G., Cely, I., Zamora, Z., Ding, J., Blencowe, M., Lang, J., Bline, A., Singh, M.,
Lusis, A.J., Yang, X., 2021. Systems toxicogenomics of prenatal low-dose BPA
exposure on liver metabolic pathways, gut microbiota, and metabolic health in mice.
Environ. Int. 146, 106260 https://doi.org/10.1016/j.envint.2020.106260.
Ehrlich, S., Calafat, A.M., Humblet, O., Smith, T., Hauser, R., 2014. Handling of thermal
receipts as a source of exposure to bisphenol A. JAMA 311, 859860. https://doi.
org/10.1001/jama.2013.283735.
Fan, S., Li, H., Zhao, R., 2020. Effects of normoxic and hypoxic conditions on the immune
response and gut microbiota of Bostrichthys sinensis. Aquaculture 525, 735336.
https://doi.org/10.1016/j.aquaculture.2020.735336.
Fu, G., Dai, J., Li, Z., Chen, F., Liu, L., Yi, L., Teng, Z., Quan, C., Zhang, L., Zhou, T.,
Donkersley, P., Song, S., Shi, Y., 2020. The role of STAT3/p53 and PI3K-Akt-mTOR
signaling pathway on DEHP-induced reproductive toxicity in pubertal male rat.
Toxicol. Appl. Pharmacol. 404, 115151 https://doi.org/10.1016/j.
taap.2020.115151.
Gao, L., Dong, Y., Lin, R., Meng, Y., Wu, F., Jia, L., 2020. The imbalance of Treg/Th17
cells induced by perinatal bisphenol A exposure is associated with activation of the
PI3K/Akt/mTOR signaling pathway in male offspring mice. Food Chem. Toxicol.
137, 111177 https://doi.org/10.1016/j.fct.2020.111177.
Gou, L., Kang, J., Dai, P., Wang, X., Li, F., Zhao, S., Zhang, M., Hua, M., Lu, Y., Zhu, Y.,
Li, Z., Chen, H., Wu, L., Li, D., Fu, X., Li, J., Shi, H., Liu, M., 2017. Ubiquitination-
decient mutations in human piwi cause male infertility by impairing histone-to
protamine exchange during spermiogenesis. Cell 169, 10901104. https://doi.org/
10.1016/j.cell.2017.04.034.
Govin, J., Lestrat, C., Caron, C., Pivot-Pajot, C., Rousseaux, S., Khochbin, S., 2006.
Histone acetylation-mediated chromatin compaction during mouse spermatogenesis.
Ernst Schering Res. Found. Workshop 57, 155172. https://doi.org/10.1007/3-540-
37633-x_9.
Health Canada, 2015. Third Report on Human Biomonitoring of Environmental
Chemicals in Canada: Results of the Canadian Health Measures Survey Cycle 3
(20122013). Available online: https://www.canada.ca/en/health-canada/services
/environmental-workplace-health/reports-publications/environmental-contamina
nts/third-report-human-biomonitoring-environmental-chemicals-canada.html.
Hu, J., Raikhel, V., Gopalakrishnan, K., Fernandez-Hernandez, H., Lambertini, L.,
Manservisi, F., Falcioni, L., Bua, L., Belpoggi, F., L. Teitelbaum, S., Chen, J., 2016.
Effect of postnatal low-dose exposure to environmental chemicals on the gut
microbiome in a rodent model. Microbiome 4, 26. https://doi.org/10.1186/s40168-
016-0173-2.
Hussain, T., Murtaza, G., Kalhoro, D.H., Kalhoro, M.S., Metwally, E., Chughtai, M.I.,
Mazhar, M.U., Khan, S.A., 2021. Relationship between gut microbiota and host-
metabolism: emphasis on hormones related to reproductive function. Anim. Nutr. 7,
110. https://doi.org/10.1016/j.aninu.2020.11.005.
Javurek, A.B., Spollen, W.G., Johnson, S.A., Bivens, N.J., Bromert, K.H., Givan, S.A.,
Rosenfeld, C.S., 2016. Effects of exposure to bisphenol A and ethinyl estradiol on the
gut microbiota of parents and their offspring in a rodent model. Gut Microbes 7,
471485. https://doi.org/10.1080/19490976.2016.1234657.
Jiang, X., Zhu, C., Li, X., Sun, J., Tian, L., Bai, W., 2018. Cyanidin-3-O-glucoside at low
doses protected against 3-Chloro-1,2-propanediol induced testis injury and improved
spermatogenesis in male rats. J. Agric. Food Chem. 66, 1267512684. https://doi.
org/10.1021/acs.jafc.8b04229.
Kong, L., Wu, Y., Hu, W., Liu, L., Xue, Y., Liang, G., 2021. Mechanisms underlying
reproductive toxicity induced by nickel nanoparticles identied by comprehensive
gene expression analysis in GC-1 spg cells. Environ. Pollut. 275, 116556 https://doi.
org/10.1016/j.envpol.2021.116556.
Lai, K., Chung, Y., Li, R., Wan, H., Wong, C.K., 2016. Bisphenol A alters gut microbiome:
comparative metagenomics analysis. Environ. Pollut. 218, 923930. https://doi.org/
10.1016/j.envpol.2016.08.039.
Li, X., Yao, Z., Yang, D., Jiang, X., Sun, J., Tian, L., Hu, J., Wu, B., Bai, W., 2020.
Cyanidin-3-O-glucoside restores spermatogenic dysfunction in cadmium-exposed
pubertal mice via histone ubiquitination and mitigating oxidative damage.
J. Hazard. Mater. 387, 121706 https://doi.org/10.1016/j.jhazmat.2019.121706.
Liu, J., Li, X., Zhou, G., Zhang, Y., Sang, Y., Wang, J., Li, Y., Ge, W., Sun, Z., Zhou, X.,
2021a. Silica nanoparticles inhibiting the differentiation of round spermatid and
chromatin remodeling of haploid period via MIWI in mice. Environ. Pollut. 284,
117446 https://doi.org/10.1016/j.envpol.2021.117446.
Liu, X., Wang, Z., Liu, F., 2021b. Chronic exposure of BPA impairs male germ cell
proliferation and induces lower sperm quality in male mice. Chemosphere 262,
127880. https://doi.org/10.1016/j.chemosphere.2020.127880.
Markle, J.G.M., Frank, D.N., Mortin-Toth, S., Robertson, C.E., Feazel, L.M., Rolle-
Kampczyk, U., von Bergen, M., McCoy, K.D., Macpherson, A.J., Danska, J.S., 2013.
Sex differences in the gut microbiome drive hormone-dependent regulation of
autoimmunity. Science 339, 10841088. https://doi.org/10.1126/science.1233521.
McLachlan, R.I., Rajpert-De, M.E., Hoei-Hansen, C.E., de Kretser, D.M., Skakkebaek, N.E.,
2007. Histological evaluation of the human testis–approaches to optimizing the
clinical value of the assessment: mini review. Hum. Reprod. 22, 216. https://doi.
org/10.1093/humrep/del279.
Molina, A., Abril, N., Morales-Prieto, N., Monterde, J., Ayala, N., Lora, A., Moyano, R.,
2018. Hypothalamic-pituitary-ovarian axis perturbation in the basis of bisphenol A
(BPA) reproductive toxicity in female zebrash (Danio rerio). Ecotoxicol. Environ.
Saf. 156, 116124. https://doi.org/10.1016/j.ecoenv.2018.03.029.
Moreira, B.P., Oliveira, P.F., Alves, M.G., 2019. Molecular mechanisms controlled by
mTOR in male reproductive system. Int. J. Mol. Sci. 20, 1633. https://doi.org/
10.3390/ijms20071633.
Murata, M., Kang, J., 2018. Bisphenol A (BPA) and cell signaling pathways. Biotechnol.
Adv. 36, 311327. https://doi.org/10.1016/j.biotechadv.2017.12.002.
Ni, Z., Sun, W., Li, R., Yang, M., Zhang, F., Chang, X., Li, W., Zhou, Z., 2021.
Fluorochloridone induces autophagy in TM4 Sertoli cells: involvement of ROS-
mediated AKT-mTOR signaling pathway. Reprod. Biol. Endocrinol. 19, 64. https://
doi.org/10.1186/s12958-021-00739-8.
Oride, A., Kanasaki, H., Tumurbaatar, T., Zolzaya, T., Okada, H., Hara, T., Kyo, S., 2020.
Effects of the fertility drugs clomiphene citrate and letrozole on kiss-1 expression in
hypothalamic kiss-1 expressing cell models. Reprod. Sci. 27, 806814. https://doi.
org/10.1007/s43032-020-00154-1.
OShaughnessy, P.J., Monteiro, A., Verhoeven, G., De Gendt, K., Abel, M.H., 2010. Effect
of FSH on testicular morphology and spermatogenesis in gonadotrophin-decient
hypogonadal mice lacking androgen receptors. Reproduction 139, 177184. https://
doi.org/10.1530/REP-09-0377.
Paris, L., Peghaire, E., Mon´
e, A., Diogon, M., Debroas, D., Delbac, F., El Alaoui, H., 2020.
Honeybee gut microbiota dysbiosis in pesticide/parasite co-exposures is mainly
induced by Nosema ceranae. J. Invertebr. Pathol. 172, 107348 https://doi.org/
10.1016/j.jip.2020.107348.
Peretz, J., Vrooman, L., Ricke, W.A., Hunt, P.A., Ehrlich, S., Hauser, R.,
Padmanabhan, V., Taylor, H.S., Swan, S.H., VandeVoort, C.A., Flaws, J.A., 2014.
Bisphenol a and reproductive health: update of experimental and human evidence,
2007-2013. Environ. Health Perspect. 122, 775786. https://doi.org/10.1289/
ehp.1307728.
Poling, M.C., Kauffman, A.S., 2013. Organizational and activational effects of sex
steroids on kisspeptin neuron development. Front. Neuroendocrin. 34, 317. https://
doi.org/10.1016/j.yfrne.2012.06.001.
Qiu, L., Chen, M., Wang, X., Qin, X., Chen, S., Qian, Y., Liu, Z., Cao, Q., Ying, Z., 2018.
Exposure to concentrated ambient PM2.5 compromises spermatogenesis in a mouse
model: Role of suppression of hypothalamus-pituitary-gonads axis. Toxicol. Sci. 162,
318326. https://doi.org/10.1093/toxsci/kfx261.
Rahman, M.S., Pang, M., 2019. Understanding the molecular mechanisms of bisphenol A
action in spermatozoa. Clin. Exp. Reprod. Med. 46, 99106. https://doi.org/
10.5653/cerm.2019.00276.
Riera, M.F., Regueira, M., Galardo, M.N., Pellizzari, E.H., Meroni, S.B., Cigorraga, S.B.,
2012R. Signal transduction pathways in FSH regulation of rat Sertoli cell
proliferation. Am. J. Physiol. Endocrinol. Metab. 302, E914E923. https://doi.org/
10.1152/ajpendo.00477.2011.
Rizzetto, L., Fava, F., Tuohy, K.M., Selmi, C., 2018. Connecting the immune system,
systemic chronic inammation and the gut microbiome: the role of sex.
J. Autoimmun. 92, 1234. https://doi.org/10.1016/j.jaut.2018.05.008.
Sharma, P., Ghanghas, P., Kaushal, N., Kaur, J., Kaur, P., 2019. Epigenetics and oxidative
stress: a twin-edged sword in spermatogenesis. Andrologia 51, e13432. https://doi.
org/10.1111/and.13432.
Singh, R.P., Shafeeque, C.M., Sharma, S.K., Pandey, N.K., Singh, R., Mohan, J.,
Kolluri, G., Saxena, M., Sharma, B., Sastry, K.V.H., Kataria, J.M., Azeez, P.A., 2015.
Bisphenol A reduces fertilizing ability and motility by compromising mitochondrial
function of sperm. Environ. Toxicol. Chem. 34, 16171622. https://doi.org/
10.1002/etc.2957.
Spaziani, M., Tarantino, C., Tahani, N., Gianfrilli, D., Sbardella, E., Lenzi, A.,
Radicioni, A.F., 2021. Hypothalamo-Pituitary axis and puberty. Mol. Cell.
Endocrinol. 520, 111094 https://doi.org/10.1016/j.mce.2020.111094.
Vandenberg, L.N., Mafni, M.V., Sonnenschein, C., Rubin, B.S., Soto, A.M., 2009.
Bisphenol-A and the great divide: a review of controversies in the eld of endocrine
disruption. Endocr. Rev. 30, 7595. https://doi.org/10.1210/er.2008-0021.
Wang, P., Luo, C., Li, Q., Chen, S., Hu, Y., 2014. Mitochondrion-mediated apoptosis is
involved in reproductive damage caused by BPA in male rats. Environ. Toxicol.
Pharmacol. 38, 10251033. https://doi.org/10.1016/j.etap.2014.10.018.
Wang, Y., Rui, M., Nie, Y., Lu, G., 2018. Inuence of gastrointestinal tract on metabolism
of bisphenol A as determined by in vitro simulated system. J. Hazard. Mater. 355,
111118. https://doi.org/10.1016/j.jhazmat.2018.05.011.
Wu, Y., Ma, J., Sun, Y., Tang, M., Kong, L., 2020. Effect and mechanism of PI3K/AKT/
mTOR signaling pathway in the apoptosis of GC-1 cells induced by nickel
nanoparticles. Chemosphere 255, 126913. https://doi.org/10.1016/j.
chemosphere.2020.126913.
Xi, W., Lee, C.K., Yeung, W.S., Giesy, J.P., Wong, M.H., Zhang, X., Hecker, M., Wong, C.
K., 2011. Effect of perinatal and postnatal bisphenol A exposure to the regulatory
circuits at the hypothalamus-pituitary-gonadal axis of CD-1 mice. Reprod. Toxicol.
31, 409417. https://doi.org/10.1016/j.reprotox.2010.12.002.
Xie, M., Bu, P., Li, F., Lan, S., Wu, H., Yuan, L., Wang, Y., 2016. Neonatal bisphenol A
exposure induces meiotic arrest and apoptosis of spermatogenic cells. Oncotarget 7,
1060610615. https://doi.org/10.18632/oncotarget.7218.
Xu, H., Shen, L., Chen, X., Ding, Y., He, J., Zhu, J., Wang, Y., Liu, X., 2016. mTOR/
P70S6K promotes spermatogonia proliferation and spermatogenesis in Sprague
Dawley rats. Reprod. Biomed. Online 32, 207217. https://doi.org/10.1016/j.
rbmo.2015.11.007.
Zhan, J., Ma, X., Liu, D., Liang, Y., Li, P., Cui, J., Zhou, Z., Wang, P., 2020. Gut
microbiome alterations induced by tributyltin exposure are associated with
R. Liu et al.
Ecotoxicology and Environmental Safety 239 (2022) 113623
13
increased body weight, impaired glucose and insulin homeostasis and endocrine
disruption in mice. Environ. Pollut. 266, 115276 https://doi.org/10.1016/j.
envpol.2020.115276.
Zhang, J., Yao, Y., Pan, J., Guo, X., Han, X., Zhou, J., Meng, X., 2020. Maternal exposure
to di-(2-ethylhexyl) phthalate (DEHP) activates the PI3K/Akt/mTOR signaling
pathway in F1 and F2 generation adult mouse testis. Exp. Cell Res. 394, 112151
https://doi.org/10.1016/j.yexcr.2020.112151.
Zhang, M., Jiang, M., Bi, Y., Zhu, H., Zhou, Z., Sha, J., 2012. Autophagy and apoptosis act
as partners to induce germ cell death after heat stress in mice. PLoS One 7, e41412.
https://doi.org/10.1371/journal.pone.0041412.
Zhao, T., Wei, Y., Wang, J., Han, L., Sun, M., Wu, Y., Shen, L., Long, C., Wu, S., Wei, G.,
2020. The gut-microbiota-testis axis mediated by the activation of the Nrf2
antioxidant pathway is related to prepuberal steroidogenesis disorders induced by
di-(2-ethylhexyl) phthalate. Environ. Sci. Pollut. Res. Int. 27, 3526135271. https://
doi.org/10.1007/s11356-020-09854-2.
Zhao, T., Tang, X., Li, D., Zhao, J., Zhou, R., Shu, F., Jia, W., Fu, W., Xia, H., Liu, G.,
2022. Prenatal exposure to environmentally relevant levels of PBDE-99 leads to
testicular dysgenesis with steroidogenesis disorders. J. Hazard. Mater. 424, 127547
https://doi.org/10.1016/j.jhazmat.2021.127547.
Abbara, A, Clarke, S.A., Dhillo, W.S, 2021. Clinical Potential of Kisspeptin in
Reproductive Health. Trends. Mol. Med. 27, 807823. https://doi.org/10.1016/j.
molmed.2021.05.008.
R. Liu et al.
... Notably, the surge in ROS contributes to oxidative stress and potentially reconfigures apoptotic cascades, manifesting as heightened cleaved Caspase 3 activity. Moreover, the activation of the phosphoinositide-3-kinase-protein kinase B pathway (PI3K/AKT) was discerned, illuminating a novel facet of BPAmediated pathogenesis [42]. ...
Article
Full-text available
Bisphenol-A (BPA), a pervasive industrial chemical used in polymer synthesis, is found in numerous consumer products including food packaging, medical devices, and resins. Detectable in a majority of the global population, BPA exposure occurs via ingestion, inhalation, and dermal routes. Extensive research has demonstrated the adverse health effects of BPA, particularly its disruption of immune and endocrine systems, along with genotoxic potential. This review focuses on the complex relationship between BPA exposure and the NOD-like receptor protein 3 (NLRP3) inflammasome, a multiprotein complex central to inflammatory disease processes. We examine how BPA induces oxidative stress through the generation of intracellular free radicals, subsequently activating NLRP3 signaling. The mechanistic details of this process are explored, including the involvement of signaling cascades such as PI3K/AKT, JAK/STAT, AMPK/mTOR, and ERK/MAPK, which are implicated in NLRP3 inflammasome activation. A key focus of this review is the wide-ranging organ toxicities associated with BPA exposure, including hepatic, renal, gastrointestinal, and cardiovascular dysfunction. We investigate the immunopathogenesis and molecular pathways driving these injuries, highlighting the interplay among BPA, oxidative stress, and the NLRP3 inflammasome. Finally, this review explores the emerging concept of targeting NLRP3 as a potential therapeutic strategy to mitigate the organ toxicities stemming from BPA exposure. This work integrates current knowledge, emphasizes complex molecular mechanisms, and promotes further research into NLRP3-targeted interventions.
... With this purpose, we collected maternal urine samples, known as one of the most important biological fluids representative of the exposure level to EDCs, and screened them for EDC concentrations, retrospectively profiling the gut microbiota composition in infants who were breastfed up to 1 year of age. Concerning the EDC-gut microbiota axis, most of the available literature was provided by studies on animal models [35][36][37][38] and, on average, these works deeply focused on a few specific EDCs. For our analyses, instead, we concomitantly considered five different chemicals, among which the BPA was also analyzed, since it appears the most studied among EDCs [39]. ...
Article
Full-text available
Endocrine disruptors (EDCs) are chemicals that interfere with the endocrine system. EDC exposure may contribute to the development of obesity, type 2 diabetes, and cardiovascular diseases by impacting the composition of an infant’s gut microbiota during the first 1000 days of life. To explore the relationship between maternal urinary levels of Bisphenol-A and phthalates (UHPLC-MS/MS), and the composition of the infant gut microbiota (16S rDNA) at age 12 months (T3) and, retrospectively, at birth (T0), 1 month (T1), and 6 months (T2), stool samples from 20 infants breastfed at least once a day were analyzed. Metataxonomic bacteria relative abundances were correlated with EDC values. Based on median Bisphenol-A levels, infants were assigned to the over-exposed group (O, n = 8) and the low-exposed group (B, n = 12). The B-group exhibited higher gut colonization of the Ruminococcus torques group genus and the O-group showed higher abundances of Erysipelatoclostridium and Bifidobacterium breve. Additionally, infants were stratified as high-risk (HR, n = 12) or low-risk (LR, n = 8) exposure to phthalates, based on the presence of at least three phthalates with concentrations exceeding the cohort median values; no differences were observed in gut microbiota composition. A retrospective analysis of gut microbiota (T0–T2) revealed a disparity in β-diversity between the O-group and the B-group. Considering T0–T3, the Linear Discriminant Effect Size indicated differences in certain microbes between the O-group vs. the B-group and the HR-group vs. the LR-group. Our findings support the potential role of microbial communities as biomarkers for high EDC exposure levels. Nevertheless, further investigations are required to deeply investigate this issue.
... Fresh fecal samples were collected from the ICR mice in DEHP-induced model at week 6 and stored at −80 • C. For bacterial diversity analysis, the V3-V4 variable region of the 16 S rRNA genes was amplified using the universal primers 338 F and 806 R [30]. ...
Article
Full-text available
Di-(2-ethylhexyl) phthalate (DEHP) is a widely used plasticizer, which can cause damage to male reproductive organs, especially the atrophy of the testis. Meanwhile, DEHP can also lead to a decrease in testicular zinc content, but the role of zinc remains unclear. This study aims to prepare oyster peptide-zinc complex (OPZC) to alleviate DEHP-induced reproductive damage in mice. OPZC was successfully obtained through electron microscopy, X-ray diffraction, and thermogravimetric analysis, with stable structure and high water-solubility. Low dose oyster peptide-zinc complex (OPZCL) significantly reduced the reproductive damage caused by DEHP in mice. Further research had shown that OPZCL restored the content of serum hormones and the activity of oxidative stress kinases to normal, while also normalizing testicular zinc and selenium levels. In addition, it also recovered the disorder of gut microbiota, reduced the proportion of Bacteroides, increased the abundance of Ligilactobacillus, and restored the proportion of Acidobacterota, Chloroflexi, and Proteobacteria. Therefore, OPZCL can relieve the reproductive damage caused by DEHP in mice by restoring testicular zinc homeostasis and the composition of intestinal microbiota, indicating that OPZCL has a potential protective effect on male reproductive health.
... For instance, a recent investigation stated that exposure to 0.5 mg/kg of BPA led to an observed increase in neuroinflammation and intestinal microbiota change via modulating the microbiota−gut−brain axis (Ni et al., 2021). Moreover, studies have elucidated that BPAinduced drastic changes in microbiota structure and short chain fatty acids (SCFAs) are likely to lead to liver damage and reproductive toxicity, which reveals that foodborne pollutants can cause body disorders through intestinal microbiota (Liu et al., 2022a;Liu et al., 2022b). Fortunately, it has been reported that anthocyanins possess intestinal regulatory capabilities. ...
Article
Full-text available
Foodborne contaminants refer to substances that are present in food and threaten food safety. Due to the progress in detection technology and the rising concerns regarding public health, there has been a surge in research focusing on the dangers posed by foodborne contaminants. These studies aim to explore and implement strategies that are both safe and efficient in mitigating the associated risks. Anthocyanins, a class of flavonoids, are abundantly present in various plant species, such as blueberries, grapes, purple sweet potatoes, cherries, mulberries, and others. Numerous epidemiological and nutritional intervention studies have provided evidence indicating that the consumption of anthocyanins through dietary intake offers a range of protective effects against the detrimental impact of foodborne contaminants. The present study aims to differentiate between two distinct subclasses of foodborne contaminants: those that are generated during the processing of food and those that originate from the surrounding environment. Furthermore, the impact of anthocyanins on foodborne contaminants was also summarized based on a review of articles published within the last 10 years. However, further investigation is warranted regarding the mechanism by which anthocyanins target foodborne contaminants, as well as the potential impact of individual variations in response. Additionally, it is important to note that there is currently a dearth of clinical research examining the efficacy of anthocyanins as an intervention for mitigating the effects of foodborne pollutants. Thus, by exploring the detoxification effect and mechanism of anthocyanins on foodborne pollutants, this review thereby provides evidence, supporting the utilization of anthocyanin‐rich diets as a means to mitigate the detrimental effects of foodborne contaminants.
... These effects depend on sex, as it can be observed in the female fetuses of rats more than in male fetuses. BPA was considered a substance affecting human health even at currently considered safe concentrations (Ruijing Liu et al, 2022). Therefore, this study was aimed to investigate the effects of BPA on the heart and its histological composition in the stage of organogenesis in the pregnant mice. ...
Article
Full-text available
The current study was aimed to identify the effect of Bisphenol A on the histology of hearts in mice during pregnancy. Thirty pregnant mice were used for the study and were divided into three groups of ten each, the first one being control group. Mice in the second and third group were given Bisphenol A orally from the sixth to eighteenth day of pregnancy. Both groups received different concentrations of Bisphenol: second group 300mg/kg and third group 450mg/kg concentration. Mice in the second group showed many histological lesions, including numerous small to moderately sized regions of myocardial degeneration and necro-sis. Myocardial fibers lost the cross striations and the nuclei were not visible. There were several extensive areas of loss or separation of myofibers characterized by increased edema. A few minor marinating neutrophilic inflamma-tory cells were seen. In contrast, mice in the third group showed myocardial necrosis and fibrin deposits between myocytes fibers. Signs of interstitial edema with significant disorganized myocardial fibers were seen. There was thickening of blood vessels accompanied by endothelial cell necrosis. An acute stage of myofiber necrosis is characterized by the presence of hyaline, and cytoplasmic eosinophilia of cardiomyocytes. Several areas of the heart showed cardiomyo-cytes with an enlarged nucleus; Fibrin thrombi and perivascular fibrosis were observed. There was endothelial cell swelling some of which were separated from the basement membrane.
Article
Full-text available
Background Fluorochloridone (FLC), a selective pyrrolidone herbicide, has been recognized as a potential endocrine disruptor and reported to induce male reproductive toxicity, but the underlying mechanism is unclear. The aim of this study was to investigate the mechanism of FLC-induced reproductive toxicity on male mice with particular emphasis on the role of autophagy in mice’ TM4 Sertoli cells. Methods Adult C57BL/6 mice were divided into one control group (0.5% sodium carboxymethyl cellulose), and four FLC-treated groups (3,15,75,375 mg/kg). The animals (ten mice per group) received gavage for 28 days. After treatment, histological analysis, sperm parameters, the microstructure of autophagy and the expression of autophagy-associated proteins in testis were evaluated. Furthermore, to explore the autophagy mechanism, TM4 Sertoli cells were treated with FLC (0,40,80,160 μM) in vitro for 24 h. Cell activity and cytoskeletal changes were measured by MTT assay and F-actin immunofluorescence staining. The formation of autophagosome, accumulation of reactive oxygen species (ROS), expression of autophagy marker proteins (LC3, Beclin-1 and P62) and AKT-related pathway proteins (AKT, mTOR) were observed. The ROS scavenger N-acetylcysteine (NAC) and AKT agonist (SC79) were used to treat TM4 cells to observe the changes of AKT-mTOR pathway and autophagy. Results In vivo, it showed that FLC exposure caused testicular injuries, abnormality in epididymal sperm. Moreover, FLC increased the formation of autophagosomes, the accumulation of LC3II/LC3I, Beclin-1 and P62 protein, which is related to the degradation of autophagy. In vitro, FLC triggered TM4 cell autophagy by increasing the formation of autophagosomes and upregulating of LC3II/LC3I, Beclin-1 and P62 levels. In addition, FLC induced ROS production and inhibited the activities of AKT and mTOR kinases. The Inhibition of AKT/mTOR signaling pathways and the activation of autophagy induced by FLC could be efficiently reversed by pretreatment of NAC. Additionally, decreased autophagy and increased cell viability were observed in TM4 cells treated with SC79 and FLC, compared with FLC alone, indicating that FLC-induced autophagy may be pro-death. Conclusion Taken together, our study provided the evidence that FLC promoted autophagy in TM4 Sertoli cells and that this process may involve ROS-mediated AKT/mTOR signaling pathways.
Article
Full-text available
It has been well recognized that interactions between the gut microbiota and host-metabolism have a proven effect on health. The gut lumen is known for harboring different bacterial communities. Microbial by-products and structural components, which are derived through the gut microbiota, generate a signaling response to maintain homeostasis. Gut microbiota is not only involved in metabolic disorders, but also participates in the regulation of reproductive hormonal function. Bacterial phyla, which are localized in the gut, allow for the metabolization of steroid hormones through the stimulation of different enzymes. Reproductive hormones such as progesterone, estrogen and testosterone play a pivotal role in the successful completion of reproductive events. Disruption in this mechanism may lead to reproductive disorders. Environmental bacteria can affect the metabolism, and degrade steroid hormones and their relevant compounds. This behavior of the bacteria can safely be implemented to eliminate steroidal compounds from a polluted environment. In this review, we summarize the metabolism of steroid hormones on the regulation of gut microbiota and vice-versa, while also examining the significant influence this process has on various events of reproductive function. Altogether, the evidence suggests that steroid hormones and gut microbiota exert a central role in the modification of host bacterial action and impact the reproductive efficiency of animals and humans.
Article
Full-text available
Bisphenol A (BPA) is an industrial plasticizer widely found in consumer products, and exposure to BPA during early development has been associated with the prevalence of various cardiometabolic diseases including obesity, metabolic syndrome, type 2 diabetes, and cardiovascular diseases. To elucidate the molecular perturbations underlying the connection of low-dose prenatal BPA exposure to cardiometabolic diseases, we conducted a multi-dimensional systems biology study assessing the liver transcriptome, gut microbial community, and diverse metabolic phenotypes in both male and female mouse offspring exposed to 5 μg/kg/day BPA during gestation. Prenatal exposure to low-dose BPA not only significantly affected liver genes involved in oxidative phosphorylation, PPAR signaling and fatty acid metabolism, but also affected the gut microbial composition in an age- and sex-dependent manner. Bacteria such as those belonging to the S24-7 and Lachnospiraceae families were correlated with offspring phenotypes, differentially expressed liver metabolic genes such as Acadl and Dgat1, and key drivers identified in our gene network modeling such as Malat1 and Apoa2. This multiomics study provides insight into the relationship between gut bacteria and host liver genes that could contribute to cardiometabolic disease risks upon low-dose BPA exposure.
Article
Testicular junctions are pivotal to male fertility and regulated by constituent proteins. Increasing evidence suggests that environmental chemicals, including bisphenol A (BPA), may impact these proteins, but whether the impacts persist for generations is not yet known. Here, we investigate the effect of BPA (a ubiquitous endocrine-disrupting chemical) on testis and sperm functions and whether the effects are transferred to subsequent generations. Male mice (F0) were exposed to corn oil (Control) or 5 or 50 mg BPA/kg body weight/day from 6 to 12 weeks of age. The F0 were mated with wild-type females to produce the first filial (F1) generation. F2 and F3 were produced using similar procedures. Our results showed that BPA doses decreased the levels of some junctional proteins partly via binding with estrogen receptors (ERα and Erβ), upregulation of p-ERK1/2, P85, p-JNK and activation of p38 mitogen-activated protein kinase signaling. Consequently, testicular histological abnormalities, disrupted spermatogenesis, decreased sperm count, and inability to fertilize eggs were observed in mice exposed to BPA. These effects were transferred to successive generations (F2), partly through DNA methylation, but mostly alleviated in F3 males. Our findings suggest that paternal exposure to chemicals promoting alteration of testicular junctional proteins and its transgenerational inheritance is a key component of the origin of male reproductive health problems.
Article
Polybrominated diphenyl ethers (PBDEs) are a widely used class of brominated flame retardants. Exposure to PBDEs could induce testicular damage in mammals, but the effects and potential mechanism of action of prenatal exposure to environmentally relevant PBDEs on testicular development remain unclear. For the in vivo study, pregnant ICR mice were exposed to environmentally relevant levels of 2,2′,4,4′,5-pentabromodiphenyl ether (PBDE-99), a major component of commercial PBDE mixtures. We found that the anogenital index and testicular organ coefficient were significantly decreased, the incidence of cryptorchidism was increased, and testicular histology was disturbed in male offspring. Transcriptomic profiling showed that steroidogenesis disorders were significant in all PBDE-99 exposure groups. The testosterone levels, expressions of testosterone regulators, and the number of CYP11A1-positive and 11β-HSD1-positive Leydig cells were significantly decreased after PBDE-99 exposure. For the in vitro study, TM3 Leydig cells were exposed to PBDE-99 at gradient concentrations. Transcriptomic profiling and validation experiments showed that PBDE-99 upregulated reactive oxygen species, activated the ERK1/2 pathway, inhibited the ubiquitination degradation pathway, and finally induced Leydig cell apoptosis. Cumulatively, these findings revealed that prenatal exposure to environmentally relevant levels of PBDE-99 leads to steroidogenesis disorders by inducing the apoptosis of Leydig cells, causing testicular dysgenesis.
Article
Kisspeptins are a family of hypothalamic neuropeptides that are essential for the regulation of reproductive physiology. Their importance in reproductive health became apparent in 2003, when loss-of-function variants in the gene encoding the kisspeptin receptor were reported to result in isolated congenital hypogonadotropic hypogonadism (CHH). It has since been ascertained that hypothalamic kisspeptin neurons regulate gonadotropin-releasing hormone (GnRH) secretion to thus stimulate the remainder of the reproductive endocrine axis. In this review, we discuss genetic variants that affect kisspeptin receptor signaling, summarize data on KISS1R agonists, and posit possible clinical uses of native and synthetic kisspeptin receptor agonists for the investigation and treatment of reproductive disorders.
Article
Researches have shown that silica nanoparticles (SiNPs) could reduce both the quantity and quality of sperm. However, the mechanism of toxicity induced by SiNPs in the male reproductive system is still unclear. In this study, male mice were randomly divided into a control group, and SiNPs treated group (20 mg/kg dose; n = 30 per group). Half of the mice per group were sacrificed on 35 days and the remaining on 50 days of the SiNPs exposure. SiNPs were found to decrease sperm count and mobility, increase the sperm abnormality rate, and damage the testes' structure. Furthermore, SiNPs decreased the protein levels of Protamine 1(PRM1) and elevated the histones' levels and suppressed the chromatin condensation of sperm. There was a significant reduction of the ubiquitinated H2A (ubH2A)/H2B (ubH2B) and RING finger protein 8 (RNF8) levels in the spermatid nucleus, while the RNF8 level in the spermatid cytoplasm increased evidently. The protein expression levels of PIWI-like protein 1(MIWI) in the late spermatids significantly increased on day 35 of SiNPs exposure. After 15 days of the withdrawal, the sperm parameters and protamine levels, and histones in the epididymal sperm were unrecovered; however, the changes in testis induced by SiNPs were recovered. Our results suggested that SiNPs could decrease the RNF8 level in the nucleus of spermatid either by upregulating of the expression of MIWI or by inhibiting its degradation. This resulted in the detention of RNF8 in the cytoplasm that maybe inhibited the RNF8-mediated ubiquitination of ubH2A and ubH2B. These events culminated in creating obstacles during the H2A and H2B removal and chromatin condensation, thereby suppressing the differentiation of round spermatids and chromatin remodeling, which compromised the sperm quality and quantity.
Article
The public around the world is increasingly concerned about male reproductive health. The impact of nickel nanoparticles (Ni NPs) on male reproductive toxicity including sperm production, motility and fertilizing capacity has been confirmed by our previous researches. In the current study of Ni NPs-inducing toxicity, the expression profiles of piRNAs and their predicted target genes associated with male infertility, were obtained. The results showed that piR-mmu-32362259 was the highest differential expression multiples in both the testis tissues of male mice and GC-1 cells similarly. Notably, piR-mmu-32362259 target gene was significantly enriched in the PI3K-AKT signaling pathway. All these results suggest that piR-mmu-32362259 may affect the occurrence and development of injury in the mouse spermatogenesis process by regulating the PI3K-AKT signaling pathway. In order to verify the result, piR-mmu-32362259 low-expression lentivirus was used to transfect GC-1 cells to establish a stable transfected cell model. The effects of piR-mmu-32362259 on the viability, cycle and apoptosis as well as related protein expression levels of GC-1 cells induced by Ni NPs were detected using CCK8, flow cytometry and western blot assay, respectively. The results showed that low expression of piR-mmu-32362259 could not only alleviate the decrease of GC-1 cell viability, affect the cell cycle and reduce the apoptosis rate, but also significantly affect the expression levels of key proteins and their downstream molecules of PI3K/AKT/mTOR signaling pathway. Collectively, our current results provide a theoretical basis for further exploring the molecular regulatory mechanism of male reproductive toxicity induced by Ni NPs.
Article
Puberty is a complex process that culminates in the acquisition of psychophysical maturity and reproductive capacity. This elaborate and fascinating process marks the end of childhood. Behind it lies a complex, genetically mediated neuroendocrine mechanism through which the gonads are activated thanks to the fine balance between central inhibitory and stimulating neuromodulators and hormones with both central and peripheral action. The onset of puberty involves the reactivation of the hypothalamic-pituitary-gonadal (HPG) axis, supported by the initial “kiss” between kisspeptin and the hypothalamic neurons that secrete GnRH (the GnRH “pulse generator”). This pulsatile production of GnRH is followed by a rise in LH and, consequently, in gonadal steroids. The onset of puberty varies naturally between individuals, and especially between males and females, in the latter of whom it is typically earlier. However, pathological variations, namely precocious and delayed puberty, are also possible. This article reviews the scientific literature on the physiological mechanisms of puberty and the main pathophysiological aspects of its onset.
Article
Tributyltin (TBT), an organotin compound once widely used in agriculture and industry, has been reported to induce obesity and endocrine disruption. Gut microbiota has a strong connection with the host’s physiology. Nevertheless, the influences of TBT exposure on gut microbiota and whether TBT-influenced gut microbiota is related to TBT-induced toxicity remains unclear. To fill these gaps, ICR (CD-1) mice were respectively exposed to TBT at NOEL (L-TBT) and tenfold NOEL (H-TBT) daily by gavage for 8 weeks in the current study. The results showed that TBT exposure significantly increased body weight as well as epididymal fat, and led to adipocyte hypertrophy, dyslipidemia and impaired glucose and insulin homeostasis in mice. Additionally, TBT exposure significantly decreased the levels of T4, T3 and testosterone in serum. Also of note, TBT exposure changed gut microbiota composition mainly by decreasing Bacteroidetes and increasing Firmicutes proportions. To confirm the role of gut microbiota in TBT-induced overweight and hormonal disorders, fecal microbiota transplantation was performed and the mice received gut microbiota from H-TBT mice had similar phenotypes with their donor mice including significant body weight and epididymal fat gain, glucose and insulin dysbiosis and hormonal disorders. These results suggested that gut microbiome altered by TBT exposure was involved in the TBT-induced increased body weight, impaired glucose and insulin homeostasis and endocrine disruption in mice, providing significant evidence and a novel perspective for better understanding the mechanism by which TBT induces toxicity.