You are on page 1of 20

Received: 24 February 2020

| Revised: 23 July 2020


| Accepted: 1 August 2020

DOI: 10.1111/jpi.12690

ORIGINAL ARTICLE

Melatonin protects mouse testes from palmitic acid-induced


lipotoxicity by attenuating oxidative stress and DNA damage in a
SIRT1-dependent manner

Dejun Xu1,2 | Lingbin Liu1 | Yongju Zhao1 | Li Yang2 | Jianyong Cheng2 |


Rongmao Hua2 | Zelin Zhang2 | Qingwang Li2

1
College of Animal Science and
Technology, Southwest University,
Abstract
Chongqing, China Palmitic acid (PA), the main component of dietary saturated fat, has been known
2
College of Animal Science and to increase in patients with obesity, and PA-induced lipotoxicity may contribute to
Technology, Northwest A&F University,
obesity-related male infertility. Melatonin has beneficial effects on reproductive pro-
Yangling, China
cesses; however, the effect and the underlying molecular mechanism of melatonin's
Correspondence involvement in PA-induced cytotoxicity in the testes are poorly understood. Our
Qingwang Li, College of Animal Science
findings showed that lipotoxicity was observed in mouse testes after long-term PA
and Technology, Northwest A&F
University, No.3 Taicheng Road, Yangling treatment and that melatonin therapy restored spermatogenesis and fertility in these
712100, China. males. Moreover, melatonin therapy suppressed PA-induced apoptosis by modu-
Email: 2015060124@nwafu.edu.cn
lating apoptosis-associated proteins such as Bcl2, Bax, C-Caspase3, C-Caspase12,
Funding information and CHOP in type B spermatogonial stem cells. Changes in the expression of endo-
Key Industry Innovation Chain of plasmic reticulum (ER) stress markers (p-IRE1, p-PERK, ATF4) and intracellular
Shaanxi Province, Grant/Award Number:
2018ZDCXL-NY-02-06; Ministry of Ca2+ levels showed that melatonin relieved PA-induced ER stress. Mechanistically,
Agriculture Transgenic Major Projects, melatonin stimulated the expression and nuclear translocation of SIRT1 through its
Grant/Award Number: 2018ZX0801013B;
receptors and prevented PA-induced ROS production and mitochondrial dysfunction
National Key Technology Support Program,
Grant/Award Number: 2015BAD03B04 via SIRT1 signaling pathway. Furthermore, melatonin promoted SIRT1-mediated
p53 deacetylation, thereby relieving G2/M arrest in response to PA-stimulated DNA
damage. Collectively, these findings indicate that melatonin protects the testes from
PA-induced lipotoxicity through the activation of SIRT1, which alleviates oxidative
stress, ER stress, mitochondrial dysfunction, and DNA damage.

KEYWORDS
DNA damage, melatonin, oxidative stress, palmitic acid, SIRT1, testis

1 | IN T RO D U C T ION this disease.2 In adipocytes, FFAs are stored and released for
energy homeostasis; however, in obese individuals, adipose
It is well known that unhealthy obesity leads to an increased tissues exhibit excessive FFAs, which lead to a myriad of
risk of male infertility.1 Although the primary pathogenesis cellular dysfunctions, which can ultimately trigger apoptotic
of obesity-associated male infertility remains unclear, exces- cell death in nonadipose cells in a process known as lipotox-
sive free fatty acids (FFAs) are recognized as a major cause of icity.3 Obesity is usually accompanied by dysregulated FFA

© 2020 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd

J Pineal Res. 2020;69:e12690.  wileyonlinelibrary.com/journal/jpi | 1 of 20


https://doi.org/10.1111/jpi.12690
2 of 20
|    XU et al.

metabolism including elevated levels of saturated fatty acids, mouse testes and type B spermatogonial stem cells (GC-1).
and in the plasma, palmitic acid (PA) is the most common Furthermore, we sought to identify the signal transduction
type of saturated FFA.4 It has been reported that levels of pathways of melatonin and the possible underlying mecha-
FFAs, including PA, are increased in patients with obesity.5-7 nisms involved in this cytoprotective action.
A clinical study also demonstrated that dietary intake of PA
was positively associated with asthenozoospermia, indicat-
ing an association between PA intake and male infertility.8 2 | M ATERIAL S AND M ETHOD S
These findings have demonstrated that elevation in PA levels
may play an important role in obesity-related male infertility. 2.1 | Chemicals
Further research indicates that excessive PA causes lipotoxic
effects, and this effect is mediated by beta-oxidation-induced Melatonin (Cat#: 73-31-4) and ER stress antagonist 4-PBA
oxidative stress.4,9 Specifically, mammalian spermatozoa (Cat#: 1716-12-7) were purchased from Sigma-Aldrich,
are especially vulnerable to reactive oxygen species (ROS), whereas melatonin receptor antagonist luzindole (Cat#:
since they are rich in polyunsaturated fatty acids, which in 117946-91-5) was purchased from Santa Cruz Biotechnology,
turn leads to male infertility.10 Therefore, it is necessary to Inc. SIRT1 antagonist EX527 (Cat#: S1541) and p38 MAPK
identify effective approaches to ameliorate PA-induced lipo- antagonist BMS-582949 (Cat#: S8124) were purchased
toxicity in the testis. from Selleck chemicals. The antibodies used in this study
N-acetyl-5-methoxytryptamine (melatonin), an endoge- were as follows: anti-melatonin receptor 2 (Cat#: 41139),
nous hormone mainly secreted by the pineal gland, is also anti-SIRT2 (Cat#: 32057), anti-SIRT3 (Cat#: 39196), anti-
synthesized in reproduction-related tissues such as ovaries Cleaved Caspase3 (Cat#: 29034), anti-Cleaved Caspase12
and testes.11,12 It has various important physiological func- (Cat#: 24209), anti-CHOP (Cat#: 40744), anti-IRE1(Cat#:
tions, including regulation of inflammation,13 apoptosis,14 45107), anti-pIRE1 (Ser724; Cat#: 13013), anti-PERK (Cat#:
and metabolism.15 Recently, much attention has been given to 33247), anti-p-PERK(Ser555; Cat#: 12887), anti-ATF4
the role of melatonin in male infertility. Melatonin has been (Cat#: 32007), anti-p38 MAPK(Cat#: 49379), anti-p-p38
shown to be involved in male reproduction by modulating MAPK (Thr180/Tyr182; Cat#: 11581), anti-Acetyl-FoxO1
steroid hormone secretion16 as well as by being involved in (Cat#: 30033), and anti-Acetyl-p53 (Cat#: HW109) were
the proliferation of spermatogenic cells.17 Moreover, mela- obtained from Signalway antibody LLC; anti-SIRT1 (Cat#:
tonin protects the testis against hyperthermia, environmental 60303-1-Ig), anti-Tubulin (Cat#: 66031-1-Ig), anti-PLZF
toxins, and drug-induced damage18-20 due to its lipophilic and (Cat#: 66672-1-Ig) were obtained from ProteinTech Group,
hydrophilic free radical scavenging characteristics.21 Indeed, Inc; anti-melatonin receptor 1 (bs-0027R), anti-ERK1/2
previous studies have demonstrated the beneficial effects of (Cat#: bs0022R), anti-p-ERK1/2 (Thr202/Tyr204; Cat#:
melatonin treatment on testicular cells under pathological bs3016R), anti-FoxO1 (Cat#: bs-2537R), anti-MnSOD
conditions; however, it is unclear whether melatonin can pre- (Cat#: bs-20668R), anti-PGC1α (Cat#: bs-1832R) were pur-
vent obesity-associated male infertility, specifically in PA- chased from Bioss; anti-Oct4 (Cat#: AF0249), anti-SOX2
induced testicular damage. (Cat#: AF8034), anti-SOX9 (Cat#: AF2329), anti-BCL2
SIRT1, a member of the conserved nicotinamide ade- (Cat#: AF6285), anti-Bax (Cat#: AF0054), anti-Nrf2 (Cat#:
nine dinucleotide (NAD+)-dependent deacetylase family, AF7623), anti-p-Nrf2 (Ser40; Cat#: AF1609), anti-Catalase
has been suggested to be involved in various biological func- (Cat#: AF0084), anti-PCNA (Cat#: AF1363), and anti-
tions, including stress resistance, differentiation, and gene Histone H3 (Cat#: AF0009) were obtained from Beyotime.
silencing based on deacetylation of substrate proteins.22,23 Unless otherwise indicated, the other chemicals were pur-
Deacetylation of forkhead transcription factors of class O chased from Sigma-Aldrich.
(FOXO) by SIRT1 is involved in oxidative stress resistance.24
Similarly, SIRT1 has been shown to deacetylate and inacti-
vate p53 in response to DNA damage.25 Interestingly, recent 2.2 | Animal experiments
studies have shown that melatonin exerts biological effects
via a SIRT1-dependent mechanism in aging, inflammation, All mice in this experiment were treated in accordance with
and embryo development.26-28 Although evidence linking the Animal Research Institute Committee guidelines of
melatonin to the role of SIRT1 has been reported, only a few Northwest A&F University, China. C57BL/6 mice (6 weeks
of the molecular mechanisms involved have been addressed. old) were housed in wire cages under conditions of constant
It would be interesting to clarify the functional link between temperature (25°C), humidity (70%), and lighting (12 hours
SIRT1 and melatonin in response to PA-induced lipotoxicity. light/12 hours dark cycle), and were fed with a standard
In the present study, we investigated whether melatonin laboratory rodent diet with water provided ad libitum during
can exert a protective effect on PA-induced cytotoxicity in this study. Melatonin was dissolved in ethanol and diluted in
XU et al.   
| 3 of 20

normal saline solution to a final concentration of 5% etha- and BMS-582949 (10 μmol/L) were used in this study. In all
nol. Palmitic acid (PA) was prepared in ethanol and diluted experiments, the concentrations of ethanol and DMSO were
in normal saline solution containing albumin (10%) for the diluted to below 1% (v/v).
PA-BSA complex solution. The male mice were randomly
assigned into three groups: The PA group was injected intra-
peritoneally with PA at a dose of 100 mg/kg/d, the PA + me- 2.5 | Histology analysis
latonin group was injected intraperitoneally with melatonin
at a dose of 10 mg/kg/d before PA treatment, and the control Testicular tissues were collected and fixed in 4% (w/v) para-
group followed the same regimen and received sham injec- formaldehyde solution. After washing with PBS, samples
tions (without melatonin and PA). After 30 days of treatment, were embedded in paraffin blocks; afterward, sections (5 μm)
the mice were sacrificed and the testicular tissues were col- were cut and stained with hematoxylin and eosin (HE). The
lected for analysis. The estrous cycles of female mice were sections were analyzed, and images were captured using an
mated to males. There before, male mice were treated with optical microscope (Nikon, Japan).
PA (100 mg/kg/d) with or without melatonin (10 mg/kg/d)
for 30 days. The pups were born approximately 22 days after
mating, and the pregnancy rates and number of offspring 2.6 | Immunofluorescence staining
were recorded and statistically analyzed.
Testicular tissues and cells were fixed using 4% paraform-
aldehyde solution. For testicular tissues, the samples were
2.3 | Assessment of sperm parameters dehydrated in ethanol at different concentrations (70%, 90%,
and 100%). After embedding in paraffin blocks, the sections
Epididymal sperm were obtained according to the method of were cut and then rehydrated (xylene, 5 minutes; ethanol,
Mohammad et al.29 Briefly, a small piece of the cauda was 100%, 95%, 70%, 5 minutes each). For immunostaining, the
cut and incubated in 1 mL of prewarmed physiological saline sections and cells were permeabilized with 0.25% (v/v) Triton
at room temperature for 5 minutes to allow the movement X-100. After blocking with QuickBlock™ blocking buffer
of spermatozoa from epididymal to fluid. Sperm concentra- (Beyotime), the slides were incubated with the primary anti-
tion was measured using Neubauer's chamber as previously bodies overnight at 4°C. The details of the antibodies used are
described.30 Sperm concentration data were expressed as listed in Table S1. The next day, the samples were incubated
106 cells/mL. Furthermore, sperm motility was assessed by with secondary antibodies at room temperature in the dark for
counting motile and nonmotile sperm and was expressed as 2 hours, followed by 30 minutes incubation with 4′,6-diamid-
the percentage of motile sperm of the total sperm count. For ino-2-phenylindole (DAPI). The stained slides were mounted
sperm morphological analyses, the cauda epididymal sperm and observed using a fluorescence microscope (Nikon). The
were spread onto slides and were stained with Giemsa stain- fluorescence intensities were analyzed with Image-Pro Plus
ing solution. Sperm with morphological abnormalities in the 6.0 software (Media Cybernetics).
head and flagellum were counted and expressed as percent-
age of the total sperm count.
2.7 | Western blot analysis

2.4 | Cell culture and treatment The treated cells were collected, and total protein was extracted
with RIPA lysis buffer containing 1 mmol/L phenylmethanesul-
The type A spermatogonial stem cells (SSCs, C18-4) were fonyl fluoride. In some experiments, the nuclear proteins were
cultured in Dulbecco's modified Eagle's medium/nutrient harvested using a Nuclear Protein Extraction Kit (Cat#: P0027;
mixture F12 (DMEM/F12) supplemented with 10% fetal bo- Beyotime). Protein concentration was then measured using a
vine serum (FBS), 2 mmol/L L-glutamine, and 1% albumin BCA protein assay kit (Beyotime). Equal amounts of protein
(for PA-BSA complex solution) at 37°C in a humidified at- (20 μg) were subjected to electrophoretic separation using
mosphere containing 5% CO2. Type B SSCs (GC-1) were a 10%-15% sodium dodecyl sulfate-polyacrylamide (SDS-
cultured in DMEM supplemented with 10% FBS, 2 mmol/L PAGE) gel and were subsequently transferred onto a nitrocellu-
L-glutamine, and 1% albumin. Leydig cells (MLTC1) were lose membrane (Beyotime). After blocking with QuickBlock™
cultured in 1640 medium supplemented with 10% FBS, Blocking Buffer (Beyotime), membranes were incubated with
2 mmol/L L-glutamine, and 1% albumin. According to pre- the primary antibodies overnight at 4°C. The details of the anti-
vious reports31-34 and viability assays, the final concentra- bodies used for Western blot assays are listed in Table S2. After
tions of PA (100 μmol/L), melatonin (1 μmol/L), EX527 washing with TBST, the membranes were incubated with sec-
(2 μmol/L), 4-PBA (250 μmol/L), luzindole (10 μmol/L), ondary antibodies at room temperature for 2 hours. The protein
4 of 20
|    XU et al.

bands were exposed using a Bio-Rad imaging system (Bio- 2.10 | Measurement of lipid uptake
Rad), and band intensities were quantified using the Quantity
One software (v. 4.52; Bio-Rad Laboratories). The fatty acid uptake assay was performed with Oil Red O so-
lution (Solarbio) and BODIPY lipid probes (Thermo Fisher).
For Oil Red O staining, cells were fixed in 4% formaldehyde
2.8 | CCK-8 and Edu assays and were stained with Oil Red O solution for 30 minutes.
After washing, the nuclei were visualized with hematoxy-
Cell counting kit-8 (CCK-8, Beyotime) was used to exam- lin staining solution and observed under a light microscope
ine cell viability. Briefly, the treated cells were seeded in (Nikon). Red indicates the lipid content. For BODIPY lipid
96-well plates and were incubated with 10 μL of CCK-8 so- staining, testicular tissues were fixed in 4% formaldehyde,
lution at 37°C for 1 hour. The optical density (OD) value and the frozen sections were permeabilized with 0.3% Triton
of each well at 450 nm wavelength was determined using X-100. Slides and cells were incubated with BODIPY lipid
a microplate reader (BioTek). For 5-ethynyl-2-deoxyuridine probes (10 μg/mL), and nuclei were stained with DAPI.
Edu assays, the proliferation ability of cells was evaluated Immediately, the stained samples were observed using a
using a BeyoClick™ EdU Cell Proliferation Kit with Alexa fluorescence microscope (Nikon). Fluorescence intensities
Fluor 594 (Cat#: C0078S; Beyotime). The procedure was were analyzed using the Image-Pro Plus 6.0 software (Media
performed according to the manufacturer's instructions. After Cybernetics).
Edu staining, cells were visualized using a fluorescence mi-
croscope (Nikon), and the percentage of Edu-positive cells
was calculated as the number of Edu-positive cells out of the 2.11 | Measurement of ROS and
total number of cells (×100). MDA production

Intracellular ROS levels were observed with the fluorescent


2.9 | Caspase3 activity and TUNEL assays dye 20,70-dichlorodihydrofluorescein diacetate (DCFH-DA,
Beyotime). Briefly, the treated cells were incubated in
Caspase3 activity assays were performed using the Caspase3 DMEM containing 10 µmol/L DCFH-DA for 30 minutes at
Activity Assay Kit (Cat#: C1115; Beyotime). Briefly, treated 37°C. After washing, the labeled cells were observed using a
cells were lysed using lysis buffer. The supernatants of the fluorescence microscope (Nikon). Malondialdehyde (MDA)
homogenate were harvested by centrifugation at 12 000 g, levels were evaluated using the Lipid Peroxidation MDA
and the protein concentration was measured using a BCA Assay Kit (Cat#: S0131M; Beyotime) based on the manu-
protein assay kit. Subsequently, cell lysates were incubated facturer's instructions. Briefly, cells were lysed with RIPA
with Ac-DEVD-pNA (2 mmol/L) at 37°C for 2 hours. After buffer, and protein concentration was determined using a
incubation, absorbance was read at 405 nm using a micro- BCA kit. Cell lysates were incubated with test solution at
plate reader (BioTek). 100°C for 15 minutes. Absorbance was then read at 532 nm
For terminal deoxynucleotidyl transferase dUTP nick-end using a microplate reader (BioTek).
labeling (TUNEL) assays, DNA damage analysis was per-
formed using a Colorimetric TUNEL Apoptosis Assay Kit
(Cat#: C1098; Beyotime). Testes and spermatogonia were 2.12 | Measurement of mitochondrial
fixed in 4% (w/v) paraformaldehyde. After embedding in par- distribution
affin blocks, the testicular sections (5 μm) were rehydrated and
washed in distilled water prior to TUNEL staining. Briefly, For mitochondrial staining, treated cells were incubated
the slides and spermatogonia were permeabilized with 0.3% in DMEM containing 100 nmol/L Mito-Tracker Green
Triton X-100 for 10 minutes. Then, the endogenous peroxi- (Beyotime) in the dark at 37°C for 30 minutes. After wash-
dase of samples was inactivated with 0.3% H2O2 in PBS for ing, stained cells were observed using a fluorescence micro-
20 minutes. Samples were incubated with the TUNEL reac- scope (Nikon). Fluorescence intensities were then analyzed
tion mixture at 37°C for 60 minutes and were stained with with Image-Pro Plus 6.0 software (Media Cybernetics).
streptavidin-HRP at room temperature for 30 minutes. The
nuclei were visualized using a hematoxylin staining solution.
Afterward, the samples were observed under a light micro- 2.13 | Flow cytometry assay
scope (Nikon): TUNEL-positive cells were brown, and nu-
clei were stained as blue. The percentage of TUNEL-positive For the cell apoptosis assay, cells were evaluated using
cells was calculated as the number of brown cells out of the an Annexin V-FITC Apoptosis Detection Kit (Cat#:
total number of cells (×100). C1062M, Beyotime). After centrifugation, treated cells
XU et al.   
| 5 of 20

were suspended in 500 μL of cold binging buffer and sub- testicular mass in the PA-treated group was smaller than that
sequently incubated in this medium containing 5 μL of in the control group, and the testis/body ratio was also signif-
Annexin V-FITC and 5 μL of PI, at room temperature for icantly reduced by PA treatment. Interestingly, a downward
10 minutes. The stained cells were detected using flow trend of testicular mass caused by PA was ameliorated in mice
cytometry (Beckman Co.) and analyzed using FlowJo injected with melatonin (Figure 1A,B). Testicular hematoxy-
Software (FlowJo, LLC.). lin-eosin (HE) staining showed no obvious histopathological
For cell cycle distribution, cells were assessed using a Cell abnormalities in the PA-treated group, whereas a larger hol-
Cycle Kit (Cat#: CA1510; Solarbio). Briefly, treated cells low tendency was observed in some PA-treated seminifer-
were fixed in 70% ethanol at 4°C for 24 hours. After washing, ous tubules compared with the control group (Figure 1C). In
samples were stained with PI containing RNase A at 37°C particular, PA induced a dramatic sperm loss, and melatonin
for 20 minutes and subsequently analyzed by flow cytometry. therapy significantly promoted spermatogenic recovery in
Data were analyzed using ModFit software (Verity Software PA-injected mice (Figure 1C,D). As expected, epididymal
House). sperm concentration and motility deteriorated in the PA in-
The mitochondrial membrane potential was evaluated jection group, which was ameliorated by pretreatment with
using a JC-1 Kit (Cat#: C2006, Beyotime). Treated cells melatonin (Table 1). Sperm morphological analyses showed
were incubated with JC-1 solution at 37°C for 20 minutes that treatment with PA caused abnormal sperm morphology
and were detected using a flow cytometry 488-nm exci- and that this abnormal morphology was significantly relieved
tation with 530-nm and 585-nm emission. Data were an- when treatment was combined with melatonin (Figure 1E,F).
alyzed using Flowjo software. The decrease in the red/ Furthermore, we mated PA- or PA/Mel-exposed male mice
green fluorescence intensity ratio indicates mitochondrial to estrous females and evaluated the fertility of these males.
depolarization. As shown in Figure 1G,H, the low pregnancy rate and re-
Intracellular Ca2+ levels were measured using Fluo-4 AM duced offspring caused by PA were significantly ameliorated
(Beyotime). Treated cells were incubated with Fluo-4 AM in the melatonin therapy group, suggesting that melatonin
(2 µmol/L) solution at 37°C for 30 minutes and were eval- can restore fertility in these PA-exposed male mice.
uated by flow cytometry. Data were analyzed using FlowJo Consistent with the histopathological findings, the im-
Software. munofluorescence assay demonstrated that the expression
ROS levels were determined by DCFH-DA staining. of PLZF, a spermatogonial stem cell (SSC) specific marker
Briefly, treated cells were incubated in DMEM containing protein, was decreased after PA treatment for 30 days, which
10 µmol/L DCFH-DA at 37°C for 30 minutes. After washing, was relieved by melatonin (Figure 1I,J). However, SOX9, a
labeled cells were evaluated using flow cytometry. Data were marker of Sertoli cells, slightly decreased after PA treatment,
analyzed by FlowJo Software. and pretreatment with melatonin had almost no significant
effect on the SOX9-positive cells in the testis (Figure S1).
The results suggested that the proliferation of SSCs was re-
2.14 | Statistical analysis covered by melatonin therapy. Moreover, endogenous plu-
ripotency genes, including Oct4 and SOX2, were examined
All data are shown as the mean ± standard error of mean for their ability to self-renew testicular cells. These data
(SEM). Statistical differences were analyzed by either showed that both Oct4 and SOX2 were decreased by PA
the two-tailed Student's t test or ANOVA followed by the treatment, which was relieved by pretreatment with mela-
Student-Newman-Keuls test between two groups using SPSS tonin (Figure 1K,L,M), suggesting that melatonin restores
20.0 software (SPSS Inc). A P value <0.05 was considered spermatogenesis in PA-treated mouse. To examine the cause
as statistically significant. All of these experiments were re- of the damaged testes, BODIPY lipid probes were used to
peated at least three times unless specified otherwise. detect lipogenesis. As shown in Figure 1N, a dramatic in-
crease in fluorescence was observed in PA-treated testes,
suggesting lipid accumulation caused by PA. When treated
3 | R E S U LTS with melatonin, lipid accumulation was obviously attenuated
(Figure 1N). As excessive lipid accumulation in nonadipose
3.1 | Melatonin prevents testicular damage tissues can result in cellular lipotoxicity, upregulation of
caused by PA TUNEL-positive cells was observed in the PA-treated sem-
iniferous tubules, which was relieved by melatonin treatment
To determine the effect of melatonin on the testes in re- (Figure 1O), indicating that melatonin can protect testicular
sponse to PA administration, we histopathologically ana- cells from PA-induced DNA damage. Taken together, these
lyzed the testes in the different treatment groups. As shown results suggest that melatonin ameliorates the testicular dam-
in Figure 1A,B, after long-term PA injection (30 days), the age caused by PA.
6 of 20
|    XU et al.
XU et al.   
| 7 of 20

F I G U R E 1 Melatonin ameliorated testicular damage caused by PA. Male mice were intraperitoneally injected with PA (100 mg/kg/d)
with or without melatonin (10 mg/kg/d) for 30 d. (A) Morphology of testes in the different groups. (B) The ratio of teste/weight in the different
groups. (C) Representative images of testicular cross sections with HE staining in the different groups. Scale bars = 100 μm. (D) The number of
spermatozoa in the tubule cross sections from different treatment groups. (E) Sperm morphology in the control (n = 8), PA (n = 6), PA + Mel
(n = 6)-treated groups. (F) The percentage of normal sperm per epididymis. (G) Pregnancy rate in the different groups. (H) The number of
offspring in the different treatment males. (I, K) Representative immunofluorescent images show the expression of PLZF, Oct4, and SOX2 in the
different groups. Scale bars = 200 μm. (J, L, M) The percentage of PLZF-positive, Oct4-positive and SOX2-positive cells was shown, respectively.
(N) Representative images of BODIPY staining in the different groups. Scale bars = 200 μm. Quantification of the BODIPY fluorescence
intensity was shown. (O) Representative images of TUNEL staining in the different groups. Brown corresponds to TUNEL-positive cells, whereas
blue corresponds to nuclei. Scale bars = 300 μm. Percentage of TUNEL-positive cells was shown. Data are representative of three independent
experiments with similar results. *P < .05; **P < .01; ***P < .001

3.2 | Melatonin suppresses PA- (Figure 2E,H,I). These findings suggest that melatonin inhib-
induced apoptosis its PA-induced cell apoptosis due to downregulation of apop-
totic proteins.
To further confirm which cell type in the testis was more
vulnerable to PA, cell line viability was assessed with the
CCK-8 assay at gradient concentrations of PA in vitro. As 3.3 | Melatonin relieves PA-induced
shown in Figure 2A, cell viability for type B SSCs (GC1) ER stress
and type A SSCs (C18-4) was more vulnerable than that of
the Leydig cells (MLTC1). In particular, the cytotoxic ef- Because of the changes in the expression of ERS-related ap-
fect of PA appeared to dramatically increase at concentra- optotic proteins, protein levels of ER stress markers were de-
tions higher than 100 μmol/L in GC-1 cells (Figure 2A). termined. PA treatment alone clearly increased p-IRE1 and
Interestingly, pretreatment with melatonin at concentrations p-PERK signals as well as ATF4 expression, suggesting that
of 0.1, 1.0, and 10.0 μmol/L significantly ameliorated the drastic ER stress occurred in PA-exposed cells (Figure 3A-
cell viability of GC-1 cells exposed to 100 μmol/L PA for D); however, sodium phenylbutyrate (4-PBA), an ER stress
24 hours, and melatonin at a concentration of 1.0 μmol/L was inhibitor, significantly decreased the phosphorylation levels
shown to be most beneficial in terms of conferring resistance of IRE1 and PERK, and ATF4 expression in PA-treated cells
to cytotoxicity (Figure 2B). Following this, a concentration (Figure 3A-D). Consistent with 4-PBA’ action, these changes
of 100 μmol/L PA and 1.0 μmol/L melatonin was used in in ER stress markers can be alleviated with the efficacy of
subsequent experiments unless otherwise stated. melatonin therapy (Figure 3A-D). ER stress could disturb
To ascertain whether PA-induced cytotoxicity occurs calcium homeostasis, which affects proper protein folding
through the apoptotic pathway, a flow cytometry assay and transport. Functionally, intracellular Ca2+ levels were de-
was carried out. As shown in Figure 2C, a significant in- tected with Fluo-4 AM staining, and flow cytometry showed
crease in apoptotic cells was observed in PA-exposed cells. that a dramatic increase in intracellular Ca2+ concentration
Compared with the control group, treatment with melatonin was observed in PA-treated cells, suggesting disturbed ER
alone did not alter the rate of apoptotic cells, whereas pre- functions caused by PA (Figure 3E). Furthermore, impaired
treatment with melatonin significantly alleviated PA-induced ER function caused by PA treatment partially recovered
apoptosis (Figure 2C). To confirm these results, the activ- when the cells were co-treated with melatonin or 4-PBA
ity of Caspase3, a critical player in apoptosis, was detected (Figure 3E). These results indicate that melatonin effectively
using a Caspase3 activity assay kit. As shown in Figure 2D, blocks PA-induced ER stress and maintains ER homeostasis.
treatment with PA significantly increased Caspase3 activ-
ity, while melatonin therapy effectively relieved this PA-
induced upregulation. Western blot analysis also showed a 3.4 | Melatonin ameliorates PA-induced
marked increase in the protein expression of cleaved forms lipotoxicity by attenuating oxidative stress
of Caspase3 in the PA-treated group, while the BCL2/Bax
ratio was decreased (Figure 2E–G). Similarly, protein ex- To further reveal the potential involvement of melatonin
pression levels of pro-apoptotic proteins were suppressed by in the protective mechanism of the testes against PA-
pretreatment with melatonin (Figure 2E–G). Given that endo- induced lipotoxicity, we next measured Nrf2 signals in the
plasmic reticulum stress (ERS) is one of the main causes of PA-injected mice, which were thought to be involved in
apoptosis, an increase of ERS-related apoptotic markers, in- resistance to oxidative stress. As shown in Figure 4A, phos-
cluding Cleaved-Caspase12 and CHOP, was observed in PA- phorylation of Nrf2 (p-Nrf2) was increased in PA-treated
treated cells, which was then relieved by melatonin therapy testes, which was partially relieved by melatonin treatment.
8 of 20
|    XU et al.

T A B L E 1 Melatonin relieved
PA + Mel
the deterioration of epididymal sperm
Parameters Control (n = 8) PA (n = 6) (n = 6)
parameters caused by PA
6 a
Concentration (10 /mL) 16.03 ± 1.37 8.89 ± 1.24c 12.96 ± 1.54b
Motility (%) 71.28 ± 2.17a 42.74 ± 1.92c 64.36 ± 2.26b
Note: Data are represented as the mean percentage ± SEM. Values within a row with different letters (a, b, c)
indicate significant differences (P < .05).

Oil Red O staining and BODIPY lipid probes showed that levels, and that melatonin therapy can relieve this stress via
PA treatment resulted in increased lipid accumulation actions that are yet-unidentified.
(Figure 4B); however, pretreatment with melatonin barely
inhibited fatty acid uptake in vitro (Figure 4B), which was
a result different from that of the testes in vivo. As cyto- 3.5 | Melatonin stimulates SIRT1 pathway
toxic accumulation of lipids in nonadipose tissues might through melatonin receptors in response to PA-
lead to cellular lipotoxicity, a significant increase in malon- induced damage
dialdehyde (MDA) levels was observed in PA-treated sper-
matogonia, which was considered to be the key mechanism We then further examined the melatonin-mediated signaling
behind ROS-induced sperm damage (Figure 4C); however, pathways. As shown in Figure 5A, melatonin receptors (MT1
melatonin pretreatment prevented the MDA production by and MT2) were observed in SSCs, Leydig cells, and Sertoli
lipid peroxidation (Figure 4C). To determine whether PA cells of the mouse testes; however, long-term PA injection
is involved in oxidative stress, a DCFH-DA probe and flow alone did not significantly change the expression of mela-
cytometry analysis were used to monitor cellular ROS. As tonin receptors. Compared with the control and PA-treated
shown in Figure 4D,E, PA treatment drastically increased groups, higher fluorescent MT1 and MT2 signals were ob-
intracellular ROS accumulation, and melatonin relieved served in testes co-treated with melatonin (Figure 5A). To
elevated ROS generation via its efficient antioxidant ac- investigate whether melatonin also promoted melatonin re-
tivity. However, there was no significant change observed ceptor expression in vitro, spermatogonial stem cell lines
in the ROS content when comparing melatonin alone and (GC-1) were exposed to various concentrations of melatonin
the control groups (Figure 4D,E). Furthermore, stress-re- (0.1, 1.0, and 10.0 μmol/L). As expected, melatonin treatment
lated extracellular signal-regulated kinase (ERK) and p38 significantly enhanced MT1 and MT2 expression levels, and
MAPK signaling pathways were investigated using Western that the most stimulatory effect of melatonin was found at
blot analysis. The data showed that no significant change 1.0 μmol/L concentration (Figure 5B). To investigate whether
in ERK1/2 signals was observed in the different treatment sirtuins are involved in the response of spermatogonia to me-
groups, while a significant activation of p38 MAPK sign- latonin treatment, protein levels of SIRT1, SIRT2, and SIRT3
aling was observed in the PA-treated cells (Figure 4F-H). were measured. Western blot analysis showed that expres-
Similarly, the p-Nrf2 levels were increased by PA admin- sion levels of SIRT1, SIRT2, and SIRT3 in PA-treated cells
istration (Figure 4F,I). Consistent with the ROS generation were significantly lower than those in the control group; how-
pattern, melatonin alone did not significantly change the ex- ever, the downregulation effect of PA treatment on SIRT2
pression levels of p-p38 MAPK and p-Nrf2 (Figure 4F,H,I). and SIRT3 expression was not recovered through melatonin
However, PA-induced stimulation was relieved by mela- therapy (Figure 5C). Interestingly, melatonin administration
tonin therapy (4F, H, I). Given that phosphorylated Nrf2 has abolished the suppressive effect of SIRT1 expression in PA-
been considered to drive its subcellular nuclear localization, treated cells (Figure 5C). Based on these results, we deter-
we set out to evaluate the correlation between melatonin mined whether the melatonin-regulated increase in SIRT1
and Nrf2 signals. As expected, immunofluorescence re- occurred through its receptors. The spermatogonia were
sults showed that an increase in the amount of nuclear Nrf2 pretreated with luzindole, a melatonin receptor antagonist,
was observed in PA-exposed cells (Figure 4J), suggesting prior to melatonin administration. As shown in Figure 5D,
that PA-induced ROS is accompanied by Nrf2 activation. the stimulatory effect of melatonin on SIRT1 expression
Interestingly, BMS-582949, a p38 MAPK antagonist, abol- was abolished by luzindole treatment of PA-exposed cells.
ished PA-induced nuclear accumulation of Nrf2, and mela- Furthermore, immunofluorescence analysis showed that MT1
tonin partially mimics this action (Figure 4J). Based on these fluorescent signals were positively correlated with SIRT1
findings, it is indicated that Nrf2 activation caused by p38 during the co-treatment of PA with melatonin (Figure 5E,F).
MAPK signaling is compensatory for the response to PA- Conversely, this relationship between melatonin and SIRT1
induced oxidative stress, but it is not enough to reduce ROS was abrogated by luzindole pretreatment (Figure 5E,F).
XU et al.   
| 9 of 20

F I G U R E 2 Melatonin relieved PA-induced apoptosis. (A) The viability of cell line was analyzed at gradient concentrations of PA treatment
for 24 h in type A SSCs (C18-4), type B SSCs (GC-1), and Leydig cells (MLTC1). (B) Effect of pretreatment with melatonin on the viability of PA
(100 μmol/L)-exposed GC-1 cells at different times. (C) Spermatogonia were pretreated with melatonin (1.0 μmol/L) for 2 h and PA (100 μmol/L)
was added for an additional 24 h. Apoptosis of spermatogonia was detected by flow cytometry analysis in the control, Mel, PA, PA + Mel-treated
groups. Percentages of the apoptotic cells were shown. (D) Caspase3 activity was shown in the different groups. (E) Western blot analysis of BCL2,
Bax, Cleaved-Caspase3, Cleaved-Caspase12, CHOP in the different groups. (F-I) The bar graph showed the quantification of Bax/BCL2, Cleaved-
Caspase3/Tubulin, Cleaved-Caspase12/Tubulin, and CHOP/Tubulin, respectively. Data are representative of three independent experiments with
similar results. *P < .05; **P < .01

Subsequently, quantitative immunofluorescence revealed a 3.6 | Melatonin relieves PA-induced


decrease of over 4-fold in the subcellular nuclear localization oxidative stress through the MT/SIRT1/FoxO1
of SIRT1 in PA-treated cells, which was restored by melatonin signaling pathways
administration (Figure 5E,G). However, melatonin-enhanced
subcellular nuclear localization of SIRT1 was abolished by To investigate how melatonin protects against PA-induced
luzindole treatment (Figure 5E,G). Collectively, these find- oxidative stress, forkhead box protein O1 (FoxO1), a tran-
ings suggest that melatonin may rely on its receptors to acti- scription factor that acts as a regulator of cell response to
vate SIRT1 pathway. oxidative stress, was analyzed in spermatogonia (GC-1). As
10 of 20
|    XU et al.

F I G U R E 3 Melatonin relieved
PA-induced endoplasmic reticulum stress
(ERS). The spermatogonia were cultured
with melatonin (1.0 μmol/L) for 2 h with or
without ERS inhibitor 4-PBA (250 μmol/L),
prior to PA treatment (100 μmol/L) for 24 h.
Cells without any treatment were used as
blank controls. (A) Western blot detection
of ERS-related proteins p-IRE1, IRE1,
p-PERK, PERK, and ATF4 in the control,
PA, PA + Mel, and PA + 4-PBA treated
groups. (B-D) The bar graph showed the
quantification of p-IRE1/IRE1, p-PERK/
PERK, and ATF4/Tubulin, respectively.
(E) Flow cytometry analysis showed
intracellular Ca2+ levels in the different
groups. Data are representative of three
independent experiments with similar
results. *P < .05; **P < .01; ***P < .001

shown in Figure 6A, a significant increase in FoxO1 acety- activation of MT/SIRT1 signals in PA-stimulated cells. To
lation was detected in PA-treated cells, which was recov- determine whether melatonin could modulate downstream
ered by melatonin therapy. Interestingly, co-incubation of SIRT1 signaling pathway, the protein expression levels of
spermatogonia with either an MT antagonist (luzindole) or antioxidant enzymes, including Catalase and MnSOD, were
a SIRT1 antagonist (EX527) abolished the downregulation investigated. Consistent with PA-induced oxidative stress,
effect of melatonin on acetylated FoxO1 (Figure 6A), sug- PA treatment resulted in low levels of Catalase and MnSOD
gesting that melatonin inhibits FoxO1 acetylation via the (Figure 6A), indicating that the cellular antioxidant system

F I G U R E 4 Melatonin ameliorated PA-induced lipotoxicity by attenuating oxidative stress. (A) Representative immunofluorescent images
showed p-Nrf2 expression in mouse testes depending on the different treatments. Scale bars = 200 μm. The fluorescence intensity of p-Nrf2
was shown from 12 testicular tissues. (B) Representative images of lipid content in spermatogonia by Oil Red O (Scale bars = 100 μm.) or
BODIPY (Scale bars = 200 μm.) staining. Quantification of the BODIPY fluorescence intensity was shown in the different groups. (C) Levels
of malondialdehyde (MDA) showed lipid peroxidation in the different groups. (D) Representative images of cellular ROS levels in the different
groups. Scale bars = 200 μm. (E) Flow cytometry analysis showed the intracellular ROS content in the different groups. (F) Protein expression
levels of p-ERK, ERK, p-p38 MAPK, p38 MAPK, and p-Nrf2 in the different groups. (G-I) The bar graph showed quantification of p-ERK/ERK,
p-p38 MAPK/p38 MAPK, and p-Nrf2/Tubulin, respectively. (J) Immunofluorescence staining showing the subcellular localization of the Nrf2
protein (red) and the nuclei stain DAPI (blue) in spermatogonia. Scale bars = 100 μm. Quantification of the colocalization efficiency between Nrf2
and DAPI in the different groups. Data are representative of three independent experiments with similar results. *P < .05; **P < .01; ***P < .001
XU et al.   
| 11 of 20
12 of 20
|    XU et al.

is damaged at a high level of ROS. However, a significant by either luzindole or EX527. These results confirmed that
upregulation of Catalase and MnSOD was observed after co- melatonin promoted SIRT1-dependent antioxidant signal-
treatment of PA with melatonin, which was then abrogated ing. Furthermore, Western blot analysis of spermatogonia
XU et al.   
| 13 of 20

F I G U R E 5 Melatonin activated SIRT1 pathway through melatonin receptors in response to PA-induced damage. (A) Representative
immunofluorescent images showed the expression of MT1 (green) and MT2 (red) in mouse testes with different treatment. Fluorescence intensity
of MT1 and MT2 was shown from 12 testicular tissues, respectively. Scale bars = 200 μm. (B) The effect of melatonin on MT1 and MT2 at
different concentrations (0.1, 1.0, 10 μmol/L). Fluorescence intensity of MT1 and MT2 was shown in spermatogonia. Scale bars = 200 μm. (C)
Protein expression levels of SIRT1, SIRT2, SIRT3 in the control, PA (100 μmol/L), and PA (100 μmol/L) + Mel (1.0 μmol/L) treated groups.
The bar graph showed the quantification of SIRT1, SIRT2, and SIRT3, respectively. (D) Western blot detection of SIRT1 in the control, Mel
(1.0 μmol/L), PA (100 μmol/L), PA (100 μmol/L) + Mel (1.0 μmol/L), PA (100 μmol/L) +Mel (1.0 μmol/L) + Luz (10 μmol/L)-treated groups. (E)
Immunofluorescence staining showing the levels of MT1 (green) and SIRT1 (Red), and the subcellular localization of the SIRT1 in spermatogonia.
The arrowhead showed colocalization efficiency between SIRT1 and the nucleus. (F) Fluorescence intensities of SIRT1 were shown. (G)
Quantification of the colocalization efficiency between SIRT1 and DAPI in the different groups. Data are representative of three independent
experiments with similar results. *P < .05; **P < .01; ***P < .001

nuclear extracts to determine the subcellular localization of Conversely, PA-induced mitochondrial impairment was
FoxO1 was performed. Consistently, compared to the control partially recovered by melatonin pretreatment, which was
group, PA administration significantly inhibited the subcel- then abolished by treatment with either luzindole or EX527
lular nuclear localization of FoxO1, whereas treatment with (Figure 7C,D). This suggests that downregulating MT/SIRT1
melatonin mostly restored nuclear FoxO1 accumulation in pathways with inhibitors prevents the protective effect of
PA-exposed cells (Figure 6B). Meanwhile, melatonin-in- melatonin against PA-induced mitochondrial damage. Given
duced nuclear accumulation of FoxO1 was completely abol- that PGC-1α is a key transcription factor that stimulates mito-
ished by either luzindole or EX527. Strikingly, a negative chondrial biogenesis, the protein expression of PGC-1α was
correlation was observed between FoxO1 acetylation and then detected. PA treatment decreased PGC-1α expression,
nuclear localization due to melatonin. This indicates that me- whereas retreatment with melatonin helped in the recovery
latonin enhances FoxO1 activation via a SIRT1-dependent of PGC-1α expression (Figure 7E,F). Meanwhile, treatment
deacetylation activity, thereby affecting downstream targets. with either luzindole or EX527 prevented the upregulation of
Similar findings were obtained, wherein the ameliorative ef- PGC-1α mediated by melatonin (Figure 7E,F). These find-
fect of melatonin on PA-induced oxidative stress was mostly ings suggest that melatonin prevents mitochondrial dysfunc-
abolished by either the MT antagonist or SIRT1 antagonist tion caused by PA in a SIRT1-dependent manner.
(Figure 6C). Taken together, these findings suggest that me-
latonin protects spermatogonia against PA-induced oxidative
stress through the activation of MT/SIRT1/FoxO1 signaling 3.8 | Melatonin recovers spermatogonia
pathways. proliferation in response to DNA damage
caused by PA through p53 deacetylation

3.7 | Melatonin prevents PA-induced We further explored how melatonin alleviates the loss
mitochondrial dysfunction in a SIRT1- of testicular SSCs caused by PA. As shown in Figure 8A,
dependent manner PA treatment increased DNA damage, as measured by the
TUNEL assay. However, co-treatment with melatonin sig-
Given that a high level of ROS may directly contribute to nificantly reduced PA-induced DNA damage, as indicated
mitochondrial dysfunction, which in turn decreases anti- by the decline of TUNEL-positive cells; this protective ef-
oxidant enzymes and exacerbates oxidative stress, we next fect was abolished by the SIRT1 inhibitor (Figure 8A).
evaluated the role of melatonin on mitochondrial function Consistent with this observation, flow cytometry analysis
in PA-induced spermatogonia. As expected, flow cytom- revealed that PA treatment drastically increased the number
etry analysis by JC-1 staining revealed a lower measurement of cells in the G2/M phase compared with the control cells,
of mitochondrial membrane potential in PA-treated cells suggesting that DNA damage caused by PA leads to cell
(Figure 7A,B), suggesting that PA impairs mitochondrial cycle arrest. However, cell cycle arrest in the G2/M phase
function. This effect was attenuated by melatonin therapy. was prevented when the spermatogonia were co-treated with
However, the protective effects of melatonin on mitochon- melatonin (Figure 8B). In addition, melatonin-induced cell
drial functional capacity were partially abolished by either division was abrogated by the SIRT1 inhibitor (Figure 8B).
luzindole or EX527, indicating that melatonin maintains In line with this, a significant inhibition of spermatogonia
mitochondrial function via MT/SIRT1 signaling pathways. proliferation was observed in PA-treated cells as observed
Furthermore, we detected the mitochondrial quantity using through Edu staining, which was then recovered by pretreat-
Mito-Tracker Green. As shown in Figure 7C,D, PA-exposed ment with melatonin (Figure 8C). Moreover, the SIRT1
cells showed decreased mitochondrial mass, as seen in the inhibitor reversed the stimulatory effects of melatonin on
decline of the Mito-Tracker Green fluorescence signals. cell proliferation (Figure 8C). To confirm these results, the
14 of 20
|    XU et al.

F I G U R E 6 Melatonin relieved PA-induced oxidative stress through MT/SIRT1/FoxO1 signaling pathways. Spermatogonia were cultured
with melatonin (1.0 μmol/L) for 2 h with or without the melatonin receptor inhibitor luzindole (10 μmol/L) or SIRT1 inhibitor EX527 (2.0 μmol/L)
before treatment with PA (100 μmol/L) for 24 h. Cells without any treatment were used as blank controls. (A) Western blot analysis showed
the protein levels of Ac-FoxO1, FoxO1, Catalase, and MnSOD in the control, PA, PA + Mel, PA + Mel +Luz, and PA + Mel +EX527-treated
groups. The bar graph showed quantification of Ac-FoxO1/FoxO1, Catalase/Tubulin, MnSOD/Tubulin, respectively. (B) Western blots of nuclear
FoxO1 protein in the different groups. The bar graph showed relative quantification of nuclear FoxO1 protein normalized to Histone H3. (C) Flow
cytometry analysis showed the intracellular ROS content in the different groups. Data are representative of three independent experiments with
similar results. Bars with different letters (a, b, and c) indicate significant differences

proliferative protein PCNA was detected by an immuno- disease.4 In particular, oxidative stress is a major cause of
fluorescence assay. Results showed that PA treatment mark- male infertility because testicular tissue has a very high rate
edly reduced the number of PCNA-positive cells, while of cell division and mitochondrial oxygen consumption as
pretreatment with melatonin increased PCNA-positive cells well as higher levels of fatty acids compared to other tis-
(Figure 8D). Interestingly, the melatonin-induced increase in sues.35 Melatonin is a well-known powerful antioxidant and
PCNA-positive cells was suppressed by the SIRT1 inhibitor is an endogenous hormone involved in multiple cellular pro-
(Figure 8D). Given the above results, we further measured cesses such as metastasis,36 proliferation,37 and apoptosis.38
the levels of acetylated p53, which was involved in cell cycle However, it is still unclear whether melatonin attenuates PA-
regulation in response to DNA damage. Western blot analy- induced damage in testicular cells. Here, we provided direct
sis showed that PA treatment drastically induced acetylated evidence, both in vivo and in vitro, that melatonin protects
p53 at a high level, whereas pretreatment with melatonin against lipotoxicity caused by PA through the attenuation of
recovered the normal levels of acetylated p53. Consistent oxidative stress. Furthermore, this study demonstrated that
with SIRT1-mediated DNA damage in spermatogonia, the the SIRT1 signaling pathway properties of melatonin are re-
downregulation effect of melatonin on acetylated p53 was sponsible for the alleviation of PA-induced lipotoxicity.
abolished by the inhibition of SIRT1 activity with EX527 In this study, long-term PA injection in vivo led to lipid
(Figure 8E). Overall, the experimental results suggest that accumulation and testicular injury, especially spermatogo-
melatonin depends on SIRT1-mediated p53 deacetylation, nial loss. Numerous studies have indicated that melatonin
which relieves G2/M arrest in response to PA-stimulated protects testicular cells against adverse conditions such as
DNA damage. hyperthermia, environmental toxins, and cytotoxic drugs.18-
20
Similarly, melatonin alleviated PA-induced testicular dam-
age and restored fertility in a mouse model. To elucidate the
4 | D IS C U SS ION mechanism underlying the beneficial effect of melatonin
in response to PA-induced lipotoxicity, type B SSCs were
In nonadipose tissue, excessive uptake of fatty acids such as utilized. ER stress is one of the main causes of apoptosis.
palmitic acid (PA) leads to elevated generation of deleterious According to our in vitro data, melatonin inhibited the ex-
lipids, impaired cellular function, and oxidative stress-related pression of pro-apoptotic proteins, particularly caspase12 and
XU et al.   
| 15 of 20

F I G U R E 7 Melatonin prevented PA-induced mitochondrial dysfunction in a SIRT1-dependent manner. Spermatogonia were cultured with
melatonin (1.0 μmol/L) for 2 h with or without melatonin receptor inhibitor luzindole (10 μmol/L) or SIRT1 inhibitor EX527 (2.0 μmol/L),
before treatment with PA (100 μmol/L) for 24 h. Cells without any treatment were used as blank controls. (A) Flow cytometry analysis showed
mitochondrial membrane potential via JC-1 staining. (B) The ratios of red/green fluorescence intensity were shown. (C) Representative images of
mitochondrial distribution in the different groups. Scale bars = 200 μm. (D) Fluorescence intensity of Mito-Tracker Green staining was shown.
(E) Western blot analysis showed PGC-1α expression in the different groups. (F) The bar graph showed the relative quantification of PGC-1α
normalized to Tubulin. Data are representative of three independent experiments with similar results. Bars with different letters (a, b, c, d) indicate
significant differences
16 of 20
|    XU et al.
XU et al.   
| 17 of 20

F I G U R E 8 Melatonin recovered proliferation in response to DNA damage caused by PA through p53 deacetylation. Spermatogonia were
treated with melatonin (1.0 μmol/L) for 2 h with or without SIRT1 inhibitor EX527 (2.0 μmol/L), before treatment with PA (100 μmol/L) for 24 h.
Cells without any treatment were used as blank controls. (A) Representative images of TUNEL staining in spermatogonia with different treatments.
Dark brown corresponds to TUNEL-positive cells, whereas blue corresponds to nuclei. Scale bars = 200 μm. The bar graph showed the percentage
of TUNEL-positive cells in the different groups. (B) Cell cycle distribution was shown by flow cytometry in the different groups. The bar graph
showed the percentage of spermatogonia in G0/G1, S, G2/M phase. (C) Representative images of Edu staining in spermatogonia under different
treatments. Scale bars = 200 μm. The bar graph showed the percentage of Edu-positive cell. (D) Representative immunofluorescent images of
PCNA expression in the different groups. Scale bars = 100 μm. The bar graph showed the percentage of PCNA-positive cells. (E) Western blot
analysis showed Ac-p53 expression in the different groups. The bar graph showed the relative quantification of Ac-p53 normalized to Tubulin.
Data are representative of three independent experiments with similar results. **P < .01; ***P < .001

CHOP, which are associated with ER stress. Subsequently, stress. Hence, we used special antagonists to investigate the
changes in ER stress markers showed that melatonin relieved anti-stress effects of melatonin and its underlying mecha-
PA-induced ER stress. This is in agreement with previous nism, and we focused on the potential involvement of SIRT1
studies confirming that melatonin can relieve spermatogonial in its regulation. Interestingly, our results show that the pro-
stem cell apoptosis by alleviating ER stress.39,40 Indeed, mel- tective effects of melatonin on PA-induced oxidative stress
atonin directly and indirectly ameliorates ER stress in mul- were abolished by SIRT1 inactivation. Moreover, previous
tiple pathologies. Melatonin directly promotes extracellular studies have found that FOXO transcription factors contrib-
secretion of insulin in INS-1E cells under induced ER stress ute to cellular oxidative stress via the SIRT1 deacetylase.49,50
conditions.41 Melatonin also functions as an anti-inflamma- Our observations are consistent with those of previous’ stud-
tory therapy via ER stress in acute pancreatitis.42 Based on ies showing that melatonin-induced SIRT1 activation inhib-
previous studies and our results, melatonin may repair the ad- ited acetylated FoxO1 drove its nuclear localization, thereby
verse effects of PA on male infertility by inhibiting ER stress. synthesizing antioxidant enzymes including Catalase and
Growing evidence has revealed that ER stress and oxida- MnSOD. Studies have shown that SIRT1 deacetylates ly-
tive stress as closely linked events play crucial roles in cel- sine residues in the FoxO1 DNA-binding domain and pro-
lular apoptosis. Published data have suggested that excessive motes nuclear retention of FoxO1, thereby upregulating
PA stimulates oxidative stress in multiple cell lines such as its transcriptional activity.51,52 These findings suggest that
HUVECs and podocytes.43,44 Likewise, Shi et al45 reported melatonin relieves PA-induced oxidative stress through the
that PA-stimulated ROS generation is accompanied by acti- SIRT1/FoxO1 signaling pathways.
vation of Nrf2 pathway. In this study, PA induced phosphor- Mitochondrial dysfunction has been considered as a major
ylated Nrf2 at a high level by activating p38 MAPK pathway, factor inducing cell death,53 specifically in testicular tissue
which is involved in the resistance to oxidative stress in PA- with high mitochondrial oxygen consumption. Strong evidence
injected mice. Interestingly, melatonin therapy did not acti- indicates that palmitate induces cell apoptosis through the ac-
vate Nrf2, but instead downregulated it by inhibiting the p38 tivation of the mitochondria-mediated apoptosis pathway.54
MAPK pathways in PA-exposed spermatogonia in the pres- Consistently, we found that PA activated the mitochondrial
ent study. In contrast, Wang et al46 reported that melatonin apoptosis pathway, including Bcl-2 inhibition and Bax promo-
activates the Nrf2 pathway in response to brain injury. These tion, as well as Caspase3 activation. Furthermore, the beneficial
raise the possibility that melatonin may alleviate the cellular effects of melatonin on mitochondrial function inhibited mito-
oxidative stress response by directly reducing ROS produc- chondrial oxidative apoptosis in the presence of PA. Indeed,
tion in spermatogonia. Subsequent experiments confirmed melatonin has been reported to prevent mitochondrial dysfunc-
that melatonin exerted beneficial effects in response to PA- tion in response to oxidative stress.55 Here, we showed that
induced oxidative stress. Sirtuins are emerging as mediators melatonin prevented mitochondrial respiration, as indicated by
of cellular response in various oxidative stress-mediated the upregulation of MMP and mitochondrial biogenesis. In ad-
pathological situations.47 Here, the two major subtypes of dition, melatonin recovered the expression of PGC-1α, which
melatonin receptors, MT1 and MT2, were present in testicu- regulates mitochondrial biogenesis and mitochondrial fatty
lar cells. Furthermore, expression and activity of SIRT1 were acid oxidation, as a substrate for SIRT1. Similar to our findings,
stimulated by melatonin therapy in response to PA admin- Gerhart-Hines et al56 demonstrated that SIRT1/PGC-1α path-
istration through its receptors, but SIRT2 and SIRT3 were ways regulate mitochondrial function and fatty acid oxidation in
not. Similar to our observations, previous reports have also muscle. However, the ameliorative effects of melatonin on mi-
shown that melatonin increased SIRT1 expression in mouse tochondrial dysfunction caused by PA were abolished by either
testicular cells,20,48 indicating a possible functional interac- an MT antagonist or a SIRT1 antagonist, indicating that mel-
tion between melatonin and SIRT1 in PA-induced oxidative atonin protects against PA-induced mitochondrial dysfunction
18 of 20
|    XU et al.

notion that melatonin protects spermatogonia against PA-


induced DNA damage and subsequently inhibits G2/M arrest
through the SIRT1/p53 signaling pathways.
In conclusion, this study demonstrated that melatonin
protected spermatogonia against PA-induced lipotoxicity
through a complex mechanism (Figure 9). Melatonin pre-
vents the PA-induced oxidative stress, ER stress, mitochon-
drial dysfunction, and DNA damage via the modulation of
FoxO1, PGC1α, and P53 in a SIRT1-dependent manner,
thereby alleviating lipotoxicity caused by PA. These findings
suggest that melatonin may be an attractive agent for attenu-
ating obesity-associated male infertility.

ACKNOWLEDGEMENTS
This work was supported by the Ministry of Agriculture
Transgenic Major Projects (2018ZX0801013B),
Key Industry Innovation Chain of Shaanxi Province
(2018ZDCXL-NY-02-06), and National Key Technology
Support Program (2015BAD03B04).

F I G U R E 9 A possible mechanism by which melatonin protected CONFLICT OF INTEREST


spermatogonia from palmitic acid-induced lipotoxicity. Melatonin The authors declare no competing financial interest.
prevented PA-induced oxidative stress through MT/SIRT1/FoxO1
signaling pathways and subsequently alleviated mitochondrial AUTHOR CONTRIBUTION
dysfunction and DNA damage caused by redox imbalance via the
Q. L. provided financial support and conceived the re-
modulation of PGC1α and P53 in a SIRT1-dependent manner, thereby
search study. D. X. performed the experiments, wrote the
inhibiting apoptotic cell death in spermatogonia. PA, palmitic acid;
manuscript. L. L. revised the manuscript. Y. Z. edited the
MT, melatonin receptor; ΔΨm, mitochondrial membrane potential
manuscript. L. Y., J. C. and R. H. conducted the animal ex-
perimentation, histological analysis and biochemical analy-
and maintains mitochondrial function by upregulating PGC-1α sis. Z. Z. analyzed the data. All authors read and approved
in a SIRT1-dependent manner. These data are in accordance the final manuscript.
with literature demonstrating the melatonin-mediated ameliora-
tion of mitochondrial dysfunction through SIRT1 activation.57 ORCID
Studies have shown that DNA damage is largely due to Qingwang Li https://orcid.org/0000-0001-7638-1565
oxidative stress which induces defective spermatozoa func-
tion.58,59 Cells have evolved mechanisms to retard prolifera- R E F E R E NC E S
tion in response to DNA damage.60 In the present study, we 1. Craig JR, Jenkins TG, Carrell DT, Hotaling JM. Obesity, male infer-
observed that PA-induced oxidative stress resulted in DNA tility, and the sperm epigenome. Fertil Steril. 2017;107(4):848-859.
damage, leading to G2/M arrest and subsequent cell death in 2. Khosrowbeygi A, Zarghami N. Fatty acid composition of human
spermatozoa and seminal plasma levels of oxidative stress bio-
spermatogonia. Melatonin has been shown to prevent DNA
markers in subfertile males. Prostaglandins Leukot Essent Fatty
damage caused by oxidative stress in several different tissue
Acids. 2007;77(2):117-121.
cell types including liver61 and brain.62 Consistent with pre- 3. Spalding K, Bernard S, Näslund E, et al. Impact of fat mass and dis-
vious reports, we found that melatonin prevented DNA dam- tribution on lipid turnover in human adipose tissue. Nat Commun.
age and promoted the proliferation of testes in response to 2017;8:e15253.
PA stimulation. It is worth noting that these beneficial effects 4. Palomer X, Pizarro-Delgado J, Barroso E, Vázquez-Carrera
were abolished by SIRT1 inactivation. Further experiments M. Palmitic and oleic acid: the yin and yang of fatty acids
revealed that PA-induced DNA damage caused acetylated in type 2 diabetes mellitus. Trends Endocrinol Metab.
2018;29(3):178-190.
p53 accumulation, whereas melatonin decreased this accu-
5. Stolba P, Kvapil M, Wichterle D, Dvorak P. Kinetics of free fatty
mulation via the deacetylase activity of SIRT1. Indeed, p53 is
acids in hypertriglyceridemia. Evidence for different types of insu-
a deacetylated substrate for SIRT1,63 which in turn becomes lin resistance. Ann N Y Acad Sci. 1993;683:373-374.
acetylated in response to DNA damage, and the acetylated 6. Manco M, Mingrone G, Greco AV, et al. Insulin resistance directly
form increased its transcriptional activity, thereby leading to correlates with increased saturated fatty acids in skeletal muscle
cell cycle arrest.64,65 These findings are consistent with the triglycerides. Metabolism. 2000;49:220-224.
XU et al.   
| 19 of 20

7. Warensjo E, Riserus U, Vessby B. Fatty acid composition of serum 27. Hardeland R. Melatonin and inflammation-Story of a double-edged
lipids predicts the development of the metabolic syndrome in men. blade. J Pineal Res. 2018;65(4):e12525.
Diabetologia. 2005;48:1999-2005. 28. Niu YJ, Zhou W, Nie ZW, Shin KT, Cui XS. Melatonin enhances
8. Eslamian G, Amirjannati N, Rashidkhani B, et al. Dietary fatty mitochondrial biogenesis and protects against rotenone-induced
acid intakes and asthenozoospermia: a case-control study. Fertil mitochondrial deficiency in early porcine embryos. J Pineal Res.
Steril. 2015;103:190-198. 2020;68(2):e12627.
9. Mancini A, Imperlini E, Nigro E, et al. Biological and nutri- 29. Mohammad KA, Mortaza A, Ali RT, et al. The effects of acryl-
tional properties of palm oil and palmitic acid: effects on health. amide on sperm parameters and membrane integrity of epididy-
Molecules. 2015;20:17339-17361. mal spermatozoa in mice. Eur J Obstet Gynecol Reprod Biol.
10. Sheweita AS, Tilmisany MA, Al-Sawaf H. Mechanisms of male 2010;153(1):52-55.
infertility: role of antioxidants. Curr Drug Metab. 2005;6:495-501. 30. Nirupama M, Devaki M, Nirupama R, Yajurvedi HN. Chronic in-
11. Frungieri MB, Calandra RS, Rossi P. Local actions of melatonin in termittent stress-induced alterations in the spermatogenesis and an-
somatic cells of the testis. Int J Mol Sci. 2017;18(6):e1170. tioxidant status of the testis are irreversible in albino rat. J Physiol
12. He Y, Deng H, Jiang Z, et al. Effects of melatonin on follicular atre- Biochem. 2013;69:59-68.
sia and granulosa cell apoptosis in the porcine. Mol Reprod Dev. 31. Li TY, Song L, Sun Y, et al. Tip60-mediated lipin 1 acetylation
2016;83(8):692-700. and ER translocation determine triacylglycerol synthesis rate. Nat
13. Mauriz JL, Collado PS, Veneroso C, Reiter RJ, González-Gallego Commun. 2018;9(1):e1916.
J. A review of the molecular aspects of melatonin’s anti–inflam- 32. González A, Alvarez-García V, Martínez-Campa C, Alonso-
matory actions: recent insights and new perspectives. J Pineal Res. González C, Cos S. Melatonin promotes differentiation of 3T3-L1
2013;54(1):1-14. fibroblasts. J Pineal Res. 2012;52(1):12-20.
14. Yang Y, Jiang S, Dong Y, et al. Melatonin prevents cell death and 33. Fang J, Yan Y, Teng X, et al. Melatonin prevents senescence of
mitochondrial dysfunction via a SIRT1-dependent mechanism canine adipose-derived mesenchymal stem cells through activating
during ischemic-stroke in mice. J Pineal Res. 2015;58(1):61-70. NRF2 and inhibiting ER stress. Aging. 2018;10(10):2954-2972.
15. Heo JI, Yoon DW, Yu JH, et al. Melatonin improves insulin resis- 34. Gräb J, Suárez I, van Gumpel E, et al. Corticosteroids inhibit
tance and hepatic steatosis through attenuation of alpha-2-HS-gly- Mycobacterium tuberculosis-induced necrotic host cell death by
coprotein. J Pineal Res. 2018;65(2):e12493. abrogating mitochondrial membrane permeability transition. Nat
16. Yu K, Deng SL, Sun TC, Li YY, Liu YX. Melatonin regulates Commun. 2019;10(1):e688.
the synthesis of steroid hormones on male reproduction: a review. 35. Asadi N, Bahmani M, Kheradmand A, Rafieian-Kopaei M. The
Molecules. 2018;23(2):e447. impact of oxidative stress on testicular function and the role
17. Niu B, Li B, Wu C, et al. Melatonin promotes goat spermatogonia of antioxidants in improving it: a review. J Clin Diagn Res.
stem cells (SSCs) proliferation by stimulating glial cell line-de- 2017;11(5):1-5.
rived neurotrophic factor (GDNF) production in Sertoli cells. 36. Song J, Kim OY. Melatonin modulates neuronal cell death induced
Oncotarget. 2016;7(47):77532-77542. by endoplasmic reticulum stress under insulin resistance condition.
18. Kim P, Zheng Y, Lv Y, et al. Melatonin protects the mouse testis Nutrients. 2017;9(6):e593.
against heat–induced damage. Mol Hum Reprod. 2020;26(2):65-79. 37. Kim MJ, Kim HK, Kim BS, Yim SV. Melatonin increases cell pro-
19. Kim SH, Lee IC, Baek HS, et al. Melatonin prevents gentamicin-in- liferation in the dentate gyrus of maternally separated rats. J Pineal
duced testicular toxicity and oxidative stress in rats. Andrologia. Res. 2004;37(3):193-197.
2014;46(9):1032-1040. 38. Hosseinzadeh A, Kamrava SK, Joghataei MT, et al. Apoptosis
20. Li B, He X, Zhuang M, et al. Melatonin ameliorates busulfan-in- signaling pathways in osteoarthritis and possible protective role of
duced spermatogonial stem cell oxidative apoptosis in mouse tes- melatonin. J Pineal Res. 2016;61(4):411-425.
tes. Antioxid Redox Signal. 2018;28(5):385-400. 39. Cui Y, Ren L, Li B, et al. Melatonin relieves busulfan-induced
21. Galano A, Castaneda-Arriaga R, Perez-Gonzalez A, Tan DX, spermatogonial stem cell apoptosis of mouse testis by inhib-
Reiter RJ. Phenolic melatonin-related compounds: their role iting endoplasmic reticulum stress. Cell Physiol Biochem.
as chemical protectors against oxidative stress. Molecules. 2017;44(6):2407-2421.
2016;21:e1442. 40. Ji YL, Wang H, Meng C, et al. Melatonin alleviates cadmium-in-
22. Alcendor RR, Gao S, Zhai P, et al. Sirt1 regulates aging and resistance duced cellular stress and germ cell apoptosis in testes. J Pineal Res.
to oxidative stress in the heart. Circ Res. 2007;100(10):1512-1521. 2012;52(1):71-79.
23. Lee YM, Shin SI, Shin KS, et al. The role of sirtuin 1 in osteoblastic 41. Yoo YM. Melatonin-mediated insulin synthesis during endoplas-
differentiation in human periodontal ligament cells. J Periodontal mic reticulum stress involves HuD expression in rat insulinoma
Res. 2011;46(6):712-721. INS-1E cells. J Pineal Res. 2013;55:207-220.
24. Nogueiras R, Habegger KM, Chaudhary N, et al. Sirtuin 1 and 42. Chen Y, Zhang J, Zhao Q, et al. Melatonin induces anti-inflamma-
sirtuin 3: physiological modulators of metabolism. Physiol Rev. tory effects to play a protective role via endoplasmic reticulum stress
2012;92(3):1479-1514. in acute pancreatitis. Cell Physiol Biochem. 2016;40(5):1094-1104.
25. Solomon JM, Pasupuleti R, Xu L, et al. Inhibition of SIRT1 cata- 43. Jiang XS, Chen XM, Wan JM, Gui HB, Ruan XZ, Du XG.
lytic activity increases p53 acetylation but does not alter cell sur- Autophagy protects against palmitic acid-induced apoptosis in
vival following DNA damage. Mol Cell Biol. 2006;26(1):28-38. podocytes in vitro. Sci Rep. 2017;7:e42764.
26. Jung-Hynes B, Reiter RJ, Ahmad N. Sirtuins, melatonin and circa- 44. Yang Q, Han L, Li J, et al. Activation of Nrf2 by phloretin attenuates
dian rhythms: building a bridge between aging and cancer. J Pineal palmitic acid-induced endothelial cell oxidative stress via AMPK-
Res. 2010;48(1):9-19. dependent signaling. J Agric Food Chem. 2019;67(1):120-131.
20 of 20
|    XU et al.

45. Shi Y, Sun Y, Sun X, et al. Up-regulation of HO-1 by Nrf2 activa- 58. Bisht S, Faiq M, Tolahunase M, Dada R. Oxidative stress and male
tion protects against palmitic acid-induced ROS increase in human infertility. Nat Rev Urol. 2017;14(8):470-485.
neuroblastoma BE(2)-M17 cells. Nutr Res. 2018;52:80-86. 59. Wang X, Sharma RK, Sikka SC, Thomas A, Falcone T, Agarwal A.
46. Wang Z, Ma C, Meng CJ, et al. Melatonin activates the Nrf2-ARE Oxidative stress is associated with increased apoptosis leading to
pathway when it protects against early brain injury in a subarach- spermatozoa DNA damage in patients with male factor infertility.
noid hemorrhage model. J Pineal Res. 2012;53(2):129-137. Fertil Steril. 2003;80(3):531-535.
47. Singh CK, Chhabra G, Ndiaye MA, Garcia-Peterson LM, Mack 60. Yao G. Modelling mammalian cellular quiescence. Interface
NJ, Ahmad N. The role of sirtuins in antioxidant and redox signal- Focus. 2014;4(3):e20130074.
ing. Antioxid Redox Signal. 2018;28(8):643-661. 61. Laothong U, Pinlaor P, Hiraku Y. Protective effect of melatonin
48. Xu G, Zhao J, Liu H, Wang J, Lu W. Melatonin inhibits apoptosis against Opisthorchis viverrini-induced oxidative and nitrosa-
and oxidative stress of mouse leydig cells via a SIRT1-dependent tive DNA damage and liver injury in hamsters. J Pineal Res.
mechanism. Molecules. 2019;24(17):e3084. 2010;49(3):271-282.
49. Brunet A, Sweeney LB, Sturgill JF, et al. Stress-dependent reg- 62. Wakatsuki A, Okatani Y, Izumiya C, Ikenoue N. Melatonin
ulation of FOXO transcription factors by the SIRT1 deacetylase. protects against ischemia and reperfusion–induced oxida-
Science. 2004;303(5666):2011-2015. tive lipid and DNA damage in fetal rat brain. J Pineal Res.
50. Kobayashi Y, Furukawa-Hibi Y, Chen C, et al. SIRT1 is critical 1999;26(3):147-152.
regulator of FOXO-mediated transcription in response to oxidative 63. Zannini L, Buscemi G, Kim JE, Fontanella E, Delia D. DBC1
stress. Int J Mol Med. 2005;16(2):237-243. phosphorylation by ATM/ATR inhibits SIRT1 deacetylase in re-
51. Frescas D, Valenti L, Accili D. Nuclear trapping of the fork- sponse to DNA damage. J Mol Cell Biol. 2012;4:294-303.
head transcription factor FoxO1 via Sirt-dependent deacetyla- 64. Barlev NA, Liu L, Chehab NH, et al. Acetylation of p53 activates
tion promotes expression of glucogenetic genes. J Biol Chem. transcription through recruitment of coactivators/histone acetyl-
2005;280:20589-20595. transferases. Mol Cell. 2001;8:1243-1254.
52. Nakae J, Cao Y, Daitoku H, et al. The LXXLL motif of murine 65. Luo J, Nikolaev AY, Imai S, et al. Negative control of p53
forkhead transcription factor FoxO1 mediates Sirt1-dependent by Sir2alpha promotes cell survival under stress. Cell.
transcriptional activity. J Clin Invest. 2006;116:2473-2483. 2001;107:137-148.
53. Marra F, Svegliati-Baroni G. Lipotoxicity and the gut-liver axis in
NASH pathogenesis. J Hepatol. 2018;68(2):280-295.
54. Yang L, Wei J, Sheng F, Li P. Attenuation of palmitic acid-induced SUPPORTING INFORMATION
lipotoxicity by chlorogenic acid through activation of SIRT1 in he-
Additional supporting information may be found online in
patocytes. Mol Nutr Food Res. 2019;63:e1801432.
55. López A, García JA, Escames G, et al. Melatonin protects the mi-
the Supporting Information section.
tochondria from oxidative damage reducing oxygen consumption,
membrane potential, and superoxide anion production. J Pineal
Res. 2009;46(2):188-198. How to cite this article: Xu D, Liu L, Zhao Y, et al.
56. Gerhart-Hines Z, Rodgers JT, Bare O, et al. Metabolic control of Melatonin protects mouse testes from palmitic acid-
muscle mitochondrial function and fatty acid oxidation through induced lipotoxicity by attenuating oxidative stress and
SIRT1/PGC1-alpha. EMBO J. 2007;26(7):1913-1923. DNA damage in a SIRT1-dependent manner. J Pineal
57. Das N, Mandala A, Naaz S, et al. Melatonin protects against lipid– Res. 2020;69:e12690. https://doi.org/10.1111/jpi.12690
induced mitochondrial dysfunction in hepatocytes and inhibits stel-
late cell activation during hepatic fibrosis in mice. J Pineal Res.
2017;62:e12404.

You might also like