Вы находитесь на странице: 1из 15

Toxicology Research

View Article Online


PAPER View Journal | View Issue

The hypothalamic–pituitary–testicular axis and


Cite this: Toxicol. Res., 2018, 7, 102
the testicular function are modulated after silver
nanoparticle exposure†
M. D. Cavallin, ‡a R. Wilk,‡a I. M. Oliveira, a N. C. S. Cardoso,a N. M. Khalil,b
c
C. A. Oliveira, M. A. Romano a and R. M. Romano *a
Published on 23 November 2017. Downloaded on 03/05/2018 03:38:16.

Silver nanoparticles (AgNPs) are widely used in industrial and medical applications and humans may be
exposed through different routes, increasing the risk of toxicity. We investigated the transcript expression
of genes involved in the regulation of the hypothalamic–pituitary–testicular (HPT) axis and the parameters
associated with sperm functionality after prepubertal exposure. AgNPs modulated the transcript
expression of genes involved in the control of the HPT axis and spermatogenesis in the groups treated
Received 31st August 2017, with lower doses, while the functional parameters related to sperm and puberty were affected in the
Accepted 22nd November 2017
groups administered higher doses. These results suggest that the HPT axis is disrupted by AgNPs during
DOI: 10.1039/c7tx00236j the prepubertal and pubertal periods, which are highly susceptible windows for the endocrine-disrupting
rsc.li/toxicology-research chemical activity.

Introduction amount of AgNPs leached by infant products (plush toys, baby


blankets, and sippy cups) and found that 43.8% of the AgNPs
Silver nanoparticles (AgNPs) have potent antimicrobial activity were leached from the spout cover of sippy cups, which rep-
against a wide range of bacteria, including highly pathogenic resents 0.93 ± 0.02 mg per kg product per day. The toxic effects
and multidrug resistant species such as Staphylococcus aureus, of silver in humans (argyria) are associated with high levels of
Salmonella typhi, Staphylococcus epidermidis and Escherichia exposure during life, but the safety limits of exposure to AgNPs
coli.1,2 For this reason, AgNPs are included in medical pro- are still under evaluation, because there is very little information
ducts such as coatings for burn wound dressings, surgical available and more risk assessment studies are necessary.16,17
devices, and bone prostheses,3–6 as well as in other consumer The toxic effects caused by inhalation,18 ingestion,19–22
products such as toothpaste, cosmetics, sprays, disinfectants, dermal contact23,24 or intravenous injection25,26 have been
deodorants, diapers, food packaging materials and water described after the in vivo administration of AgNPs in animal
purifiers.7–11 Currently, there are approximately 442 different models. These toxic effects include the inhibition of mitochon-
products that contain AgNPs in their chemical composition.12 drial activity,27,28 cytotoxicity,29 genotoxicity30 and the pro-
Therefore, humans may be exposed to AgNPs present in duction of reactive oxygen species, which causes oxidative
everyday nano-functionalized consumer products.13 Recently, stress in the central nervous system.31,32
it was demonstrated that AgNP-impregnated toothbrushes Regarding reproductive toxicity, in vitro exposure of mam-
release silver, which may cause human and environmental malian spermatogonial stem cells to AgNPs damages the cell
exposure to AgNPs.14 Furthermore, Quadros et al.15 tested the membrane, induces apoptosis and necrosis,33 and promotes
the decline in cellular proliferation.34 Rabbits exposed intra-
venously to AgNPs have decreased sperm motility and
a
Laboratory of Reproductive Toxicology, Department of Pharmacy, State University of increased production of reactive oxygen species.35
Centro-Oeste, Rua Simeao Camargo Varela de Sa, 03, 85040-080, Parana, Brazil.
Furthermore, studies in rats showed that oral exposure to
E-mail: romano@unicentro.br
b
Laboratory of Nanotechnology, Department of Pharmacy, State University of Centro- AgNPs reduces sperm production22 and integrity, compro-
Oeste, Rua Simeao Camargo Varela de Sa, 03, 85040-080, Parana, Brazil mises the integrity of plasma and acrosome membranes,
c
Laboratory of Hormonal Dosages, Department of Animal Reproduction, Faculty of reduces the mitochondrial activity in the mid-piece of the
Veterinary Medicine, University of Sao Paulo, Av. Prof. Dr. Orlando Marques de sperm, and alters the reproductive behavior.21
Paiva, 87, 05508-270, Sao Paulo, Brazil
Healthy spermatozoa are the final product of a complex
† Electronic supplementary information (ESI) available. See DOI: 10.1039/
c7tx00236j process controlled by the hypothalamic–pituitary–testicular
‡ These authors contributed equally to the work. (HPT) axis. In this context, this study sought to investigate the

102 | Toxicol. Res., 2018, 7, 102–116 This journal is © The Royal Society of Chemistry 2018
View Article Online

Toxicology Research Paper

molecular mechanisms involved in the dose–response effects Results


of four dosages of AgNPs (1.875, 3.75, 7.5 or 15 µg AgNPs per
kg) on the regulation of the HPT axis and the testicular func- Characterization of AgNPs
tion, using prepubertal male Wistar rats as an experimental Prior to initiating the experimental protocol it was necessary to
model. During the entire experimental period (from postnatal determine the AgNP diameter and polydispersity indexes
day (PND) 23 through PND60) the animals were weighed daily before and after dilution of the commercial solution. The
to follow their growth, and upon reaching puberty the weight dilution did not alter the particle diameter, which was esti-
and age were recorded. mated to be 79.5 ± 11 nm and the polydispersity index
The secretion of gonadotropins is finely regulated by the measured was 0.575 ± 118. The stability of AgNPs in simulated
hypothalamus, where the pulse frequency of gonadotropin- gastric fluid is presented in ESI Table 1.†
releasing hormone (GnRH) controls the differential expression
of LH and FSH subunit genes by the gonadotrophs in the pitu- The effects of prepubertal AgNP exposure on the body growth,
itary.36,37 Thus, the hypothalamus was evaluated for the tran- onset of puberty and reproductive organ weights
script expression (mRNA) of gonadotropin releasing hormone 1 Following AgNP prepubertal exposure the rats were routinely
Published on 23 November 2017. Downloaded on 03/05/2018 03:38:16.

(Gnrh1); and the pituitary was evaluated for the transcript weighed to monitor body growth, and had no effect on any of
expression of gonadotropin releasing hormone receptor the treated groups, as compared to controls (ESI Fig. 1†).
(Gnrhr), luteinizing hormone beta polypeptide (Lhb), and fol- However, the onset of puberty was delayed in the groups
licle stimulating hormone beta subunit (Fshb). Additionally, the receiving AgNP dosages 7.5 and 15 µg kg−1, when compared to
serum LH and FSH concentrations were measured in the blood. the controls (Fig. 1). Furthermore, the weights of the testis, epi-
The negative feedback mechanism was investigated in both didymis segments, seminal vesicle and seminal fluid were not
the hypothalamus and pituitary by the transcript expression of affected by any of the treatment dosages (Table 1).
the androgen receptor (Ar), estrogen receptor 1 (Esr1) and
estrogen receptor 2 (Esr2), since these receptors may be Effects of AgNPs on gonadotropin production
involved in controlling the synthesis and release of GnRH and
Towards the goal of understanding how AgNPs modulate gon-
LH.38 The transcript expression of inhibin B (Inhbb) in the
adotropin production, we first examined the transcript
testis was also analyzed, because of its participation in the
expression of Gnrh in the hypothalamus, since this can influ-
negative feedback of FSH.39
ence the expressions of LH and FSH in the pituitary. As shown
Once in the blood, LH stimulates the synthesis of testoster-
in Fig. 2A, only the 3.75 µg AgNP per kg-treated group dis-
one by binding to the luteinizing hormone/choriogonadotro-
played an increase in Gnrh expression, as compared to con-
pin receptor (LHCGR) in the Leydig cells in the testis.40 Thus,
trols; however, the transcript expression of Gnrhr in the pitu-
the testes were evaluated for the transcript expression of Lhcgr
itary was unaffected (Fig. 2B).
and the serum testosterone concentration was measured in the
While the Lhb transcript expression in the pituitary did not
blood. In the Sertoli cells, testosterone binds to the androgen
change in any of the treated groups (Fig. 3A), the serum con-
receptor (AR) or is converted into estradiol by the aromatase
centrations of LH increased in the 3.75 µg AgNP per kg-treated
(CYP19A1) enzyme, both of these events are related to the sper-
group, as compared to the controls (Fig. 3B). There were also
matogenesis process.40 Therefore, the relative transcript
no significant changes in the Fshb transcript expression in the
expressions of Ar and Cyp19a1 were evaluated in the testis and
pituitary (Fig. 3C), yet the serum concentrations of FSH
the serum estradiol concentration was measured in the blood.
increased in the 1.875 µg AgNP per kg-treated group, when
Spermatogenesis events take place in the Sertoli cells, and
compared to the controls (Fig. 3D).
are mainly influenced by the action of FSH, which binds to the
follicle stimulating hormone receptor (FSHR) to perform its
action.40 Estradiol is another important hormone for the sper-
matogenesis process, interacting with estrogen receptors
(ESR1 and ESR2) located in the nucleus and cytoplasm or with
G protein-coupled estrogen receptor 1 (GPER) in the plasma
membrane, which is crucial for the regulation of proteins
involved in the function of the testis.41 In this sense, we evalu-
ated the transcript expression of Fshr, Esr1, Esr2 and Gper in
the testis.
Finally, the sperm production, sperm reserves and sperm
transit time in the epididymis segments were evaluated. The
sperm functionality was assessed by the verification of acro-
Fig. 1 Age at puberty onset for rats exposed to 0 (control), 15, 7.5, 3.75
some integrity, plasma membrane integrity, sperm defects and
or 1.875 µg of AgNPs per kg BW. The values are expressed as median
mitochondrial activity, since these methods more accurately and interquartile ranges (Kruskal–Wallis followed by the post-test of
indicate subfertility and infertility, which reflect the ability of Dunn), *P < 0.05 vs. control, n = 12 per group. AgNPs: silver nano-
the sperm to fertilize the oocyte.42–44 particles; BW: body weight.

This journal is © The Royal Society of Chemistry 2018 Toxicol. Res., 2018, 7, 102–116 | 103
View Article Online

Paper Toxicology Research

Table 1 The testis, epididymis, seminal vesicle and seminal fluid relative weights in animals exposed to silver nanoparticles (AgNPs)

AgNPs (µg kg−1)


Organ weight
(mg per 100 g BW) 0 1.875 3.75 7.5 15

Testis 487 ± 18 468 ± 7 494 ± 14 488 ± 10 524 ± 10


Caput + corpus epididymis 69 ± 1.5 64 ± 1.4 73 ± 1.5 74 ± 2 67 ± 2
Cauda epididymis 44.5 ± 1.2 47 ± 0.8 49 ± 1.2 42 ± 0.8 44 ± 1.5
Seminal vesicle undrained 232 ± 9 208 ± 14 223 ± 8 214 ± 11 198 ± 6
Seminal fluid 123 ± 8 139 ± 16 134 ± 7 125 ± 9 109 ± 6

Data were subjected to analysis of variance and are expressed as mean ± standard error of the mean; n = 12 animals per group.

per kg-treated groups, when compared to the controls


(Fig. 4C).
Published on 23 November 2017. Downloaded on 03/05/2018 03:38:16.

In the pituitary, the Ar transcript expression was decreased


in the 1.875, 3.75 and 7.5 µg AgNP per kg-treated groups
(Fig. 5A), the Esr1 transcript expression was not altered in any
of the treated groups (Fig. 5B) and the Esr2 transcript
expression was decreased in the 1.875, 3.75 and 7.5 µg AgNP
per kg-treated groups, as compared to the controls (Fig. 5C).
In the testis, the Inhbb transcript expression was found to
be increased only in the 7.5 µg AgNP per kg-treated group,
when compared to controls (Fig. 6).

Effects of AgNPs on testicular steroidogenesis


Having established that the onset of puberty and that the tran-
script expression levels of some of the molecular components
involved in gonadotropin synthesis and secretion were modu-
lated, we then investigated the effects of AgNP exposure on tes-
ticular steroidogenesis. The Lhcgr transcript expression in the
testis was not altered in any of the groups treated with AgNPs
(Fig. 7A). An increase in the serum concentrations of testoster-
one was observed in the 1.875 µg AgNP per kg-treated group,
Fig. 2 Transcript expression of Gnrh (A) and Gnrhr (B) for rats exposed when compared to the controls (Fig. 7B). Interestingly, the
to 0 (control), 15, 7.5, 3.75 or 1.875 µg of AgNPs per kg BW. The Rpl19 estradiol serum concentration was increased in the 7.5 AgNP
was used as an internal control. The values are expressed as the mean ± per kg-treated group (Fig. 7D), despite not being able to detect
S.E.M. (ANOVA followed by the post-test of Dunnett), *P < 0.05 vs.
a significant increase ( p = 0.08) in the transcript expression of
control, n = 12 per group. Gnrh: Gonadotropin releasing hormone;
Gnrhr: gonadotropin releasing hormone receptor; Rpl19: ribosomal
aromatase (Cyp19a1) for this group, when compared to the
protein L19; AgNPs: silver nanoparticles; BW: body weight; SEM: stan- controls (Fig. 7C).
dard error of the mean.
Effects of AgNPs on the expression of genes related to
spermatogenesis
Spermatogenesis relies heavily on the action of FSH and the
Effects of AgNPs on the expression of genes controlling the estrogen receptors; thus the transcript expression levels were
negative feedback mechanisms in the hypothalamic–pituitary– measured. The Ar transcript expression was reduced in the
testicular axis and estradiol receptors 1.875 and 3.75 µg AgNP per kg-treated groups compared to the
Next we investigated molecular components of the HPT axis control (Fig. 8A), whereas the Fshr transcript expression in the
that are involved in the negative feedback mechanism regulat- testis was not altered (Fig. 8B). In relation to the estradiol
ing the synthesis and secretion of GnRH, LH and FSH. In the receptors, the transcript expression of Gper (Fig. 9A) was
hypothalamus, the Ar transcript expression was not altered in reduced in the 1.875 µg AgNP per kg-treated group, and the
any of the groups treated with AgNPs (Fig. 4A). However, the transcript expression of Esr1 was reduced in the 1.875 and
Esr1 transcript expression was decreased in the 7.5 µg AgNP 3.75 µg AgNP per kg-treated groups, when compared to the
per kg-treated group (Fig. 4B), and the Esr2 transcript controls (Fig. 9B). On the other hand, the expression of Esr2
expression was decreased in the 1.875, 3.75 and 7.5 µg AgNP was not affected by AgNP treatment (Fig. 9C).

104 | Toxicol. Res., 2018, 7, 102–116 This journal is © The Royal Society of Chemistry 2018
View Article Online

Toxicology Research Paper


Published on 23 November 2017. Downloaded on 03/05/2018 03:38:16.

Fig. 3 Pituitary transcript expression of Lhb (A) and Fshb (C), serum concentrations of LH (B) and FSH (D) for rats exposed to 0 (control), 15, 7.5,
3.75 or 1.875 µg of AgNPs per kg BW. The Rpl19 was used as an internal control. The values are expressed as the mean ± S.E.M. (ANOVA followed by
the post-test of Dunnett), *P < 0.05 vs. control, n = 12 per group. Lhb: luteinizing hormone beta polypeptide; Fshb: follicle stimulating hormone beta
subunit; LH: lutropin; FSH: follitropin; Rpl19: ribosomal protein L19; AgNPs: silver nanoparticles; BW: body weight; SEM: standard error of the mean.

Effects of AgNPs on the sperm production, sperm reserves and nants, which may increase the incidence of male reproductive
sperm transit time disorders.45 In the present study, we investigated the mole-
With regard to sperm parameters, none of the AgNP dosages, cular mechanisms involved in the dose–response effects of
used in this study, had an effect on sperm production four dosages of AgNPs (1.875, 3.75, 7.5 or 15 µg AgNPs per kg)
(Table 2). However, the sperm reserves (Fig. 10A) and the on the regulation of the HPT axis and the testicular function
sperm transit time (Fig. 10C) through the caput epididymis using prepubertal male Wistar rats as an experimental model.
were decreased in the 1.875 µg AgNP per kg-treated group, The mean size of silver nanoparticles estimated by dynamic
when compared to controls. On the other hand, the sperm light scattering was 79.5 nm, possibly due to differences in the
reserves (Fig. 10B) and the sperm transit time (Fig. 10D) in the evaluation method (the manufacturer used transmission elec-
cauda epididymis were not affected by AgNP treatment. tron microscopy7,46). In the simulated gastric fluid, the mean
particle size of AgNPs increased with time in the low pH
Effect of AgNPs on the sperm functionality environment of the stomach. These data are similar to those
Finally, we examined sperm functionality parameters, which reported by Pinďáková et al. (2017).47 In the simulated intesti-
may impact the ability of the sperm to fertilize the oocyte. The nal fluid, the AgNPs did not present this behavior, remaining
acrosome integrity was reduced in the 15 µg AgNP per kg- dispersed in the solution.47 After oral exposure, the liver, the
treated group, when compared to controls (Fig. 11A); however, kidney and the blood present higher levels of AgNPs, which
the plasma membrane integrity and the sperm morphology consistently decline in the subsequent weeks.48 Nevertheless,
were unchanged in all of the AgNP dosages utilized in this in the testis and brain the levels are still elevated.48–50 This
study (Fig. 11B and C). The mitochondrial activity was reduced may contribute to the development of the toxic effects
in the 7.5 and 15 µg AgNP per kg-treated groups, as compared observed in this study.
to controls (Fig. 11D). The central nervous system is the main initiator of puberty
in rats. A network not yet fully understood controls the GnRH
neurons, in which before puberty their activity is predomi-
Discussion nantly inhibitory and after puberty predominantly excitatory.51
Prior to puberty, a pulsatile release of GnRH is increased and
Reproductive physiology involves complex biological processes stimulatory events lead to the release of LH and FSH from pitu-
that may be sensitive to environmental chemical contami- itary, which will perform their action in the testicular cells.51–53

This journal is © The Royal Society of Chemistry 2018 Toxicol. Res., 2018, 7, 102–116 | 105
View Article Online

Paper Toxicology Research


Published on 23 November 2017. Downloaded on 03/05/2018 03:38:16.

Fig. 5 Pituitary transcript expression of Ar (A), Esr1 (B) and Esr2 (C) for
Fig. 4 Hypothalamic transcript expression of Ar (A), Esr1 (B) and Esr2
rats exposed to 0 (control), 15, 7.5, 3.75 or 1.875 µg of AgNPs per kg BW.
(C) for rats exposed to 0 (control), 15, 7.5, 3.75 or 1.875 µg of AgNPs per
The Rpl19 was used as an internal control. The values are expressed as
kg BW. The Rpl19 was used as an internal control. The values are
the mean ± S.E.M. (ANOVA followed by the post-test of Dunnett), *P <
expressed as the mean ± S.E.M. (ANOVA followed by the post-test of
0.05, **P < 0.01 and ***P < 0.001 vs. control, n = 12 per group. Ar:
Dunnett), *P < 0.05 and **P < 0.01 vs.control, n = 12 per group. Ar:
androgen receptor 1; Esr1: estradiol receptor 1; Esr2: estradiol receptor
androgen receptor 1; Esr1: estradiol receptor 1; Esr2: estradiol receptor
2; Rpl19: ribosomal protein L19; AgNPs: silver nanoparticles; BW: body
2; Rpl19: ribosomal protein L19; AgNPs: silver nanoparticles; BW: body
weight; SEM: standard error of the mean.
weight; SEM: standard error of the mean.

The action of FSH in the Sertoli cells helps to coordinate the occurred during the prepubertal period. In addition to these
onset of puberty and progression, but the androgens play a events, peripheral insulin growth factor 1 (IGF-1) stimulates
more relevant role in the process.52 The balanopreputial separ- GnRH secretion, which correlates positively with body growth
ation is a good indicator of the progress of puberty in rats and the progress of puberty.51 In this study, the body develop-
because there is a positive correlation between the rise in the ment was not affected by AgNP oral exposure during the prepu-
serum concentrations of testosterone at the beginning of bertal period. In fact, this parameter is affected by oral exposure
puberty and the externalization of the penis from the glans.54 to AgNPs only at higher dosages (125 mg kg−1) and after longer
Exposure to chemicals with estrogenic or antiandrogenic effects periods of treatment in male rats.57
can delay the onset of puberty in males.55,56 The delayed After puberty, the hypothalamic neurons release GnRH in a
puberty in the rats, treated with 7.5 and 15 µg AgNPs per kg, pulse frequency that stimulates the expression of Lhb and Fshb
observed in this study suggests that hormonal imbalances and the secretion of LH and FSH by the gonadotrophs in the

106 | Toxicol. Res., 2018, 7, 102–116 This journal is © The Royal Society of Chemistry 2018
View Article Online

Toxicology Research Paper

and the transcript expression of Esr2 is reduced (1.875, 3.75


and 7.5 µg AgNPs per kg), which could suggest that there is a
disruption in estradiol signaling in the hypothalamus.
Furthermore, AgNP treatment (7.5 µg AgNPs per kg) also
reduced the transcript expression of Esr1 in the hypothalamus.
The transcript expressions of gonadotropins (Lhb and Fshb)
were not affected by treatment, but the serum concentrations
of LH and FSH were increased (3.75 and 1.875 µg AgNPs per
kg, respectively). This increase may be related to mechanisms
involved in the posttranscriptional regulation, such as the half-
life of mRNA in the cytoplasm, the translational rate of
Fig. 6 Testicular transcript expression of Inhbb for rats exposed to 0 mRNA,62 or through altered gonadotropin secretion.63
(control), 15, 7.5, 3.75 or 1.875 µg of AgNPs per kg BW. The Rpl19 was The negative feedback mechanism in the hypothalamus
used as an internal control. The values are expressed as the mean ±
and pituitary is mediated by testosterone binding to the AR,
S.E.M. (ANOVA followed by the post-test of Dunnett), *P < 0.05 vs.
Published on 23 November 2017. Downloaded on 03/05/2018 03:38:16.

control, n = 12 per group. Inhbb: Inhibin beta B subunit; Rpl19: which represses the synthesis and release of LH, while the
ribosomal protein L19; AgNPs: silver nanoparticles; BW: body weight; negative feedback of FSH is mediated, in the pituitary, by the
SEM: standard error of the mean. action of inhibin b, which represses its synthesis and
release.39 The transcript expression of Ar is reduced in the
pituitary of rats (1.875, 3.75 and 7.5 µg AgNPs per kg), weaken-
pituitary.36,37 The regulation of GnRH-producing neurons is ing the negative feedback mechanism controlling the synthesis
still under investigation,58 but is suggestive that estradiol and secretion of LH. The transcript expression of Inhbb is only
receptors mediate their activity.59 Estradiol binds to ESR2 acti- elevated in the 7.5 µg AgNPs per kg group. These molecular
vating the transcriptional complex, which promotes the tran- mechanisms are unclear, but the disruption in the LH syn-
scription of the GnRH gene,60 while ESR1 activation is the pre- thesis and secretion may perturb the fine regulation of FSH,
dominant mechanism involved in the estradiol-mediated sup- since it is produced by the same pituitary cells.
pression of GnRH synthesis/release.61 In this study, the tran- In the testis, LH stimulates the synthesis of testosterone by
script expression of Gnrh1 is increased (3.75 µg AgNPs per kg) the Leydig cells.40 The exposure to AgNPs increased the serum

Fig. 7 Testicular transcript expression of Lhcgr (A) and Cyp19a1 (C), serum concentrations of testosterone (B) and estradiol (D), for rats exposed to
0 (control), 15, 7.5, 3.75 or 1.875 µg of AgNPs per kg BW. The Rpl19 was used as an internal control. The values are expressed as the mean ± S.E.M.
(ANOVA followed by the post-test of Dunnett), *P < 0.05 and **P < 0.01 vs. control, n = 12 per group. Lhcgr: luteinizing hormone/choriogonadotro-
pin receptor; Cyp19a1: aromatase; Rpl19: ribosomal protein L19; AgNPs: silver nanoparticles; BW: body weight; SEM: standard error of the mean.

This journal is © The Royal Society of Chemistry 2018 Toxicol. Res., 2018, 7, 102–116 | 107
View Article Online

Paper Toxicology Research


Published on 23 November 2017. Downloaded on 03/05/2018 03:38:16.

Fig. 8 Testicular transcript expression of Ar (A) and Fshr (B) for rats
exposed to 0 (control), 15, 7.5, 3.75 or 1.875 µg of AgNPs per kg BW. The
Rpl19 was used as an internal control. The values are expressed as the
mean ± SEM; ANOVA followed by the post-test of Dunnett; *P < 0.05
and ***P < 0.001 vs. control, n = 12 animals per group; Ar: androgen
receptor; Fshr: follicle stimulating hormone receptor; Rpl19: ribosomal
protein L19; AgNPs: silver nanoparticles; BW: body weight; SEM: stan-
dard error of the mean.

concentrations of testosterone (1.875 µg AgNPs per kg). In pre-


vious studies using higher doses (15, 30 or 50 µg AgNPs per
Fig. 9 Testicular transcript expression of Gper (A), Esr1 (B) and Esr2 (C)
kg) this parameter was not affected.21,22
for rats exposed to 0 (control), 15, 7.5, 3.75 or 1.875 µg of AgNPs per kg
Testosterone binds to the AR in the Sertoli cells40 and plays BW. The Rpl19 was used as internal control. The values are expressed as
a key role in the maintenance of spermatogenesis, participat- the mean ± S.E.M. (ANOVA followed by the post-test of Dunnett), *P <
ing in the maintenance of the blood–testis barrier, as well as 0.05 and **P < 0.01 vs. control, n = 12 per group. Gper: estrogen recep-
the adhesion and release of sperm from the Sertoli cells.64,65 tor 1 coupled to protein G; Esr1: estradiol receptor 1; Esr2: estradiol
receptor 2; Rpl19: ribosomal protein L19; AgNPs: silver nanoparticles;
The transcript expression of Ar was reduced (1.875 and 3.75 µg
BW: body weight; SEM: standard error of the mean.
AgNPs per kg).
Normal sexual development and reproduction in mammals
depends on the balance between androgens and estrogens. In
the testis, maintaining this balance relies on endocrine and The estradiol interacts with estrogen receptors located in
paracrine factors, but it is also related to the activity of aroma- the nucleus and cytoplasm (ESR1 and ESR2) or in the plasma
tase, an enzyme complex located in the endoplasmic reticulum membrane (GPER), which is crucial for the regulation of pro-
that ensures the conversion of androgen into estrogen.66 teins involved in the function of the testis.41 In fact, ESR1 and
Testicular estradiol is produced in the testis from the aromatiza- ESR2 knockout mice exhibit compromised spermatogenesis.69
tion of testosterone by the aromatase cytochrome P450 family Furthermore, ESR1 and ESR2 were found in the testis of adult
19 subfamily A member 1 enzyme, encoded by the gene and immature rats and were also expressed in co-culture of
CYP19A1.67 In the present study, serum estradiol concentrations Sertoli cells obtained from mice at 15 days of age.70
were increased (7.5 µg AgNPs per kg), whereas Cyp19a1 Additionally, GPER has been detected in the extranuclear
expression was not altered in any treated group. Interestingly, regions of Sertoli cells and can regulate the expression of
the estradiol serum concentrations and aromatase expression genes involved in cell apoptosis.71 It has also been shown that
were not affected in the 1.875 µg AgNP per kg-treated group ESR1 and ESR2 are involved in the control of cell proliferation
(increased testosterone), which may indicate a protective mecha- and differentiation by modulating enhancers and inhibitors of
nism avoiding higher and more deleterious estradiol levels.68 the cell cycle.72 Additionally, ESR1 is necessary for the viability

108 | Toxicol. Res., 2018, 7, 102–116 This journal is © The Royal Society of Chemistry 2018
View Article Online

Toxicology Research Paper

Table 2 Sperm production in animals exposed to silver nanoparticles (AgNPs)

AgNPs (µg kg−1)

Sperm production 0 1.875 3.75 7.5 15

Total
×106 per testis 97.1 ± 3.5 98.3 ± 8.5 92.7 ± 8.3 98.1 ± 10.0 99.4 ± 7.2
×106 per g testis 69.0 ± 2.9 70.4 ± 6.0 64.8 ± 5.9 69.8 ± 7.5 65.6 ± 5.4
Daily
×106 per testis 15.9 ± 0.5 16.1 ± 1.4 15.2 ± 1.3 16.1 ± 1.6 16.3 ± 1.1
×106 per g testis 11.3 ± 0.4 11.5 ± 0.9 10.6 ± 0.9 11.4 ± 1.2 10.8 ± 0.8

Data were subjected to analysis of variance and are expressed as the mean ± standard error of the mean; n = 12 animals per group.
Published on 23 November 2017. Downloaded on 03/05/2018 03:38:16.

Fig. 10 Sperm reserves (A and B) and sperm transit time (C and D) in epididymis segments for rats exposed to 0 (control), 15, 7.5, 3.75 or 1.875 µg
of AgNPs per kg BW. The values are expressed as the mean ± SEM; ANOVA followed by the post-test of Dunnett; *P < 0.05 vs. control, n = 12 per
group. AgNPs: silver nanoparticles; BW: body weight; SEM: standard error of the mean.

of germ cells and is involved in chromatin condensation and through the epididymis segments, the sperm released from
DNA integrity of spermatozoa.73 In this study, the expression the lumen of seminiferous tubules undergoes maturation and
of the transcripts of Gper was decreased in the group receiving acquires motility and the ability to fertilize oocytes.74 The
the lowest dosage (1.875 µg AgNPs per kg), whereas the tran- sperm transit time through the epididymis segments is due to
scripts of Esr1 were decreased in the groups treated with 1.875 the movement of the fluid secreted by the Sertoli cells, which
and 3.75 µg AgNPs per kg. As a consequence of these altera- results from the contractile activity of the smooth muscle epi-
tions a reduction in the sperm production could occur, but it didymis, which is controlled by androgen action and the auto-
was surprisingly not affected. A possible explanation is that nomic nervous system.75 In this study, the reduced sperm
the spermatic cycle could be compromised, because Sleiman reserves and the sperm transit time may be due to increased
et al.22 only observed a reduction in the sperm production serum concentrations of testosterone which could stimulate
thirty-seven days after AgNP exposure. In this sense, the com- the contraction of the smooth muscle of the epididymis; as a
promised spermatogenesis observed in these groups exposed result these increased contractions could contribute to the
to lower doses of AgNPs should not be disregarded. observed decrease in sperm transit time.
Furthermore, the sperm reserves and the sperm transit Next, the sperm collected from the cauda epididymis was
time in caput and corpus epididymis were reduced in the subjected to the functional analysis of acrosome integrity,
1.875 µg AgNP per kg-treated group. During the transit plasma membrane integrity, mitochondrial activity and sperm

This journal is © The Royal Society of Chemistry 2018 Toxicol. Res., 2018, 7, 102–116 | 109
View Article Online

Paper Toxicology Research


Published on 23 November 2017. Downloaded on 03/05/2018 03:38:16.

Fig. 11 Frequency of acrosome integrity (A), plasma membrane integrity (B), defects (C) and mitochondrial activity (D) of the sperm for rats exposed
to 0 (control), 15, 7.5, 3.75 or 1.875 µg of AgNPs per kg BW. The values are expressed as the mean ± SEM; ANOVA followed by the post-test of
Dunnett; *P < 0.05 vs. control, n = 12 per group. AgNPs: silver nanoparticles; BW: body weight; SEM: standard error of the mean.

morphology. In this study, the sperm morphology and the chondrial activity, decreased hypothalamic expression of
plasma membrane integrity were not affected. The acrosome Esr1, increased expression of Inhbb and elevated serum con-
integrity was reduced (15 µg AgNPs per kg) and the mitochon- centrations of estradiol. The group receiving 15 µg AgNPs per
drial activity was reduced (7.5 and 15 µg AgNPs per kg). The kg presented delayed puberty onset and decreased sperm
acrosome plays a fundamental role in the fertilization process functionality, yet all of the other parameters were unaffected.
and the reduction in its integrity may compromise the ability These differences may be related to the levels of dose–
to fertilize.76 Mitochondria are located in the middle piece of response in the HPT axis, and help in understanding the
the sperm, which produces energy to move the flagellum and meaning of the modulation of the studied genes and the
propel the spermatozoa through the genital tract. In this way, a functional parameters. At lower doses (1.875 and 3.75 µg
reduction in its function may compromise sperm mobility and AgNPs per kg), the modulation of the genes in the HPT axis
consequently hinder fertilization.77 Alterations in the group avoid disturbances in its function. At higher dose (15 µg
receiving the higher dose (15 µg AgNPs per kg) were previously AgNPs per kg), the genes are not being modulated but the
demonstrated by our group,21 and this was included in this function is compromised.
study to represent the highest dosage in the dose–response Finally, humans may be exposed to AgNPs by everyday con-
curve. Thus, it was possible to infer a LOAEL (Lowest Observed sumer products which are nano-functionalized13 and for
Adverse Effect Level) of 7.5 µg AgNPs per kg and a NOAEL (No which the safety limits of exposure are still under evaluation.16
Observed Adverse Effect Level) of 3.75 µg AgNPs per kg for It is known that AgNP-impregnated toothbrushes release Ag,
spermatic function. which may cause human and environmental exposure to
Taking all these results together, some differences were AgNps.14 Quadros et al.15 tested the amount of AgNPs leached
observed among the AgNP-treated groups. The groups receiv- by infant products ( plush toys, baby blankets, and sippy cups),
ing 1.875 and 3.75 µg AgNPs per kg did not present any and found that 43.8% of the AgNPs leached from a sippy cup
alteration in the functional parameters analyzed, but only in (spout cover), representing 0.93 ± 0.02 mg kg−1 product per
the analysis at the molecular level. The group receiving day.15 The same study was also evaluated by probabilistic
1.875 µg AgNPs per kg was the only group to display methods to assess the relative uncertainty and potential risks
increased serum concentrations of FSH and testosterone. The of exposure of infants to AgNPs and it was concluded that the
group receiving 3.75 µg AgNPs per kg was the only group to leach from these infant products exhibited a minimal risk.78
present increased transcript expression of Gnrh1 and The relevance of human exposure to low doses of AgNPs is a
increased LH serum concentrations. The group receiving controversial issue and in this sense it is necessary to perform
7.5 µg AgNPs per kg had delayed puberty onset, reduced mito- more risk assessment studies.

110 | Toxicol. Res., 2018, 7, 102–116 This journal is © The Royal Society of Chemistry 2018
View Article Online

Toxicology Research Paper

Experimental determine the age at the onset of puberty.54 This evaluation


was performed on PND33 and was continued every other day
Materials and methods throughout the balanopreputial separation period by gentle
Characterization of AgNPs. A suspension of silver nano- tissue manipulation. The animals were weighed every other
particles 60 nm in diameter was purchased from Sigma- day from PND23 to PND60 to determine body growth.
Aldrich (catalog number 730815, Sigma-Aldrich Co., Seelze, Tissue collection. At PND60, animals were euthanized by
Germany). Dilution with water was performed just before the decapitation under deep anesthesia with ketamine and xyla-
suspension was administered to the animals according to our zine. Blood was collected by cardiac puncture, immediately
previous study.21 To evaluate whether diluting the AgNP solu- centrifuged at 205g (II Excelsa BL 206, Sao Paulo, SP, Brazil)
tion affected the nanoparticle stability, we analyzed the mean for 15 min, and the serum was maintained at −80 °C until
particle size and polydispersity index by dynamic light scatter- further analysis of hormonal dosages of LH, FSH, testosterone
ing (DSL) (BIC 90 plus – Brookhaven Instruments Corp., USA) and estradiol. The hypothalamus, the pituitary and testes were
at a scattering angle of 90° and a temperature of 25 °C, with immediately excised, frozen in liquid nitrogen and kept at
the help of ZetaPlus Particle Sizing Software Version 5.34 −80 °C until further analysis of gene expression by RT-qPCR.
Published on 23 November 2017. Downloaded on 03/05/2018 03:38:16.

(Brookhaven Instruments Corp., USA). The particle size of The testes, epididymis (caput, corpus and cauda), seminal
AgNPs was evaluated in simulated gastric fluid. For this, vesicles and seminal fluid were weighed and converted to mg
AgNPs were diluted in ultra-purified water or acidic conditions of tissue per 100 mg of body weight.
(simulated gastric fluid, SGF, without pepsin, pH 1.2; USP Assessment of sperm functionality. The sperm was collected
201279) to a final concentration of 0.004 mg mL−1 and kept in from the cauda epididymis by making an incision with a
a platform shaker at 200 rpm and 37 °C. The samples were scalpel blade and squeezing out the fluid. Twenty microliters
taken after 0, 30, 60 and 120 minutes and analyzed by DLS of the drained fluid containing spermatozoa were suspended
under the same conditions as those described above. in 200 µl of a seminal extender (328.8 mM Tris, 91.3 mM fruc-
Experimental design. The experimental design was based on tose, and 115.8 mM citric acid) at 37 °C. The samples were
a protocol suggested by Stoker et al.56 from the Endocrine then immediately subjected to functional analysis of the acro-
Disrupting Screening and Testing Advisory Committee some integrity, plasma membrane integrity, mitochondrial
(EDSTAC) and includes the evaluation of endocrine-disrupting activity and sperm morphology, as previously described.21,80
chemical effects in prepuberty and puberty. Sixty newly The acrosome integrity was evaluated by using a single-
weaned male Wistar rats (Rattus norvegicus var. albinus) were stain solution containing 1% (w/v) rose bengal (cat. number
collected from pregnant female rats that had been monitored 330000, Sigma-Aldrich Co, MO, USA), 1% (w/v) fast green FCF
from the 17th day of pregnancy to determine the exact day of (cat. number F7252, Sigma-Aldrich Co, MO, USA), and 40%
birth. On postnatal day 4 (PND4), 10 litters were culled to 8 ethanol in 200 mM disodium phosphate buffer containing
pups (4 males/4 females) per female and kept at this size until 100 mM citric acid at pH 7.2. Five microliters of each sample
weaning (PND21). On PND23, the male offspring were divided were incubated with 5 µl of the single-stain solution for 1 min
into five groups containing 12 animals each, and received at 37 °C. The total volume (10 µl) was then pipetted onto a
either 0 (control), 1.875, 3.75, 7.5 or 15 μg of AgNPs per kg of slide and smeared with another slide, air-dried at 37 °C and
body weight (BW) from PND23 to PND60. The highest dose of analyzed by light microscopy (1000×). Two hundred spermato-
AgNPs for the dose–response curve used in this study was zoa were counted per slide and were classified as having intact
based on the experimental toxicological values for the lowest or damaged acrosomes.
observable adverse effect level (LOAEL) for sperm parameters The plasma membrane integrity was evaluated using eosin–
(15 μg kg−1),21 and corresponds to a dose that is 2000-fold nigrosin stain [1% eosin Y (Sigma-Aldrich, MO, USA) and 10%
lower than the NOAEL for liver toxicity.17,57 The AgNP water nigrosin (Sigma-Aldrich, MO, USA) in distilled water]. Five
suspension was administered every other day at a volume of microliters of each sample were mixed with 5 µl of the stain,
0.25 ml per 100 g BW by gavage. The control group received and the total volume was pipetted onto a slide, smeared with
only water. During all experiments, the animals were kept in another slide, and air-dried at room temperature. Two
groups of four, housed in polypropylene cages (43 × 43 × hundred spermatozoa were counted per slide by light
20 cm) with a 5 cm layer of wood shavings, fed commercial microscopy (1000×) and were classified as having intact or
feed (Nuvilab CR-1, Nuvital, PR, Brazil) and were allowed damaged membranes.
access to water ad libitum, under a 12 : 12 hour dark/light cycle The mitochondrial activity was evaluated by measuring the
in a temperature-controlled room (23 ± 1 °C). All procedures enzyme activity of cytochrome c oxidase in the intermembrane
were performed in accordance with the Conselho Nacional de space of the spermatozoa using DAB (3,3′-diaminobenzidine,
Controle de Experimentação Animal (CONCEA) and were Sigma-Aldrich, MO, USA). DAB was diluted in PBS (2.7 mM
approved by the Universidade Estadual do Centro-Oeste KCl, 137 mM NaCl, 8 mM NaHPO4, and 1.4 mM KPO4, pH 7.4)
Ethical Committee for Animal Research ( protocol 013/2015). to a final concentration of 1 mg ml−1 and frozen until use. Ten
Puberty and body growth. An evaluation of the separation of microliters of each sample were incubated with 300 µl of a
the preputial membrane and the externalization of the glands DAB solution pre-heated to 37 °C for 1 h at 37 °C in the dark.
of the penis (balanopreputial separation, PPS) was used to Ten microliters of each sample were pipetted onto a slide,

This journal is © The Royal Society of Chemistry 2018 Toxicol. Res., 2018, 7, 102–116 | 111
View Article Online

Paper Toxicology Research

smeared with another slide, and air-dried at 37 °C in the dark. following cycling conditions: 50 °C (2 min), 95 °C (2 min) and
The slide samples were then incubated for 10 min in a fixative 40 cycles of 95 °C (15 s) and 60 °C (30 s). At the end of the reac-
solution containing 10% formaldehyde (F8775, Sigma-Aldrich tion, a melting curve was generated and analyzed to confirm
Co, MO, USA) in PBS and allowed to air-dry at 37 °C in the the specificity of the amplification. The average cycle threshold
dark. Two hundred spermatozoa from each sample were ana- (Ct) was automatically determined using StepOne Software
lyzed by phase-contrast microscopy at a magnification of 400× v2.3 (Applied Biosystems), and quantification was performed
and then classified based on the degree of staining of the using the 2−ΔΔCt method, as previously described.84 The
intermediate piece as follows: DAB I (completely stained), DAB primer pairs are shown in ESI Table 2.†
II (>50%), DAB III (<50%) and DAB IV (unstained). Hormone measurements. Estradiol and testosterone serum
The sperm morphology was evaluated as a wet preparation concentrations were analyzed by colorimetric competitive
by phase contrast microscopy (400× magnification) in diluted enzyme immunoassay using a commercial Testosterone ELISA
samples in buffered formalin–saline solution (34.72 mM kit and a 17β-estradiol ELISA kit (Enzo Life Sciences Inc.,
Na2HPO4·2H2O, 18.68 mM KH2PO4, 92.4 mM NaCl, and 12.5% Farmingdale, NY, USA). The serum LH and FSH concentrations
(v/v) formaldehyde). were determined by a chemiluminescent immunoassay using
Published on 23 November 2017. Downloaded on 03/05/2018 03:38:16.

Sperm production, sperm reserves and sperm transit time. Luminex xMAP technology (Milliplex MAP rat pituitary panel;
The sperm production, reserves and transit time were deter- Billerica, MA, USA). The intra-assay coefficients of variation
mined as previously detailed by Romano et al.,81 and accord- were <10% for testosterone, <8% for estradiol, <6% for LH and
ing to the method described by Robb et al.82 Briefly, the testes <4.5% for FSH.
were weighed, the tunica albuginea was removed and the testi- Statistical analysis. The variables were first subjected to the
cular parenchyma was homogenized in 5 ml of a saline solu- Kolmogorov–Smirnov test for normality and the Bartlett test
tion containing 0.5% Triton X-100, followed by sonication at for homoscedasticity. The analysis of body growth was per-
12 kHz for 30 seconds (Ultrasonic homogenizer DES500; formed by multi-way analysis of variance for repeated
Unique, Brazil), and the resistant spermatids were counted measures (MANOVA) using a general linear model (GLM). The
using a hemocytometer. Daily sperm production was obtained day of PPS was compared among the groups using nonpara-
by dividing the sperm production by 6.1 [spermatids counted metric analyses with the Kruskal–Wallis method followed by
in this method represent 48% of one cycle of the seminiferous the post hoc Dunn test. All other parameters were analyzed by
epithelium which has a duration of 12.75 days for rats (12.75 × ANOVA followed by the post-test of Dunnett, comparing treated
0.48 = 6.1)]. Sperm reserves were calculated from epididymis groups only with the control group. Serum concentrations of
segments (caput, corpus and cauda) that were weighed and estradiol and the Cyp19a1 relative expression were compared
minced, homogenized in 5 mL of 0.9% NaCl containing 0.5% by Pearson’s correlation. All analyses were performed with
Triton X-100 followed by sonication at 12 kHz for 30 seconds Statistica 7.0 (StatSoft Inc., Tulsa, OK). Statistical differences
(Ultrasonic homogenizer DES500; Unique, Brazil) and the were considered significant when P was less than 0.05.
spermatozoa were counted using a hemocytometer. Sperm
transit time was calculated by dividing the number of sperma-
tozoa in each segment by the daily sperm production of the Conclusions
corresponding testis.
Reverse transcription followed by real-time quantitative PCR In this study, it was observed that AgNPs modulate the
(RT-qPCR). The hypothalamus, the pituitary and testes were expression of genes related to the control of the HPT axis and
pulverized in liquid nitrogen and total RNA was extracted by spermatogenesis with repercussions in the production of
the guanidine–phenol–chloroform method,83 using Trizol serum testosterone and estradiol concentrations only in the
reagent (Life Technologies, Carlsbad, USA) according to the group receiving the lowest dosage of AgNPs, while functional
manufacturer’s instructions. The hypothalamus was evaluated alterations of the sperm were observed in the group receiving
for the transcript expression of Gnrh1, Ar, Esr1 and Esr2 rela- the higher dosages. This is suggestive of a disruption of the
tive to the expression of the ribosomal protein L19 (Rpl19). HTP axis by AgNPs during the prepubertal and pubertal
The pituitary was evaluated for the transcript expression of periods, which are the most susceptible windows for the endo-
Gnrhr, Lhb, Fshb, Ar, Esr1 and Esr2 relative to the expression of crine-disrupting chemical activity.
Rpl19. The testes were evaluated for the transcript expression
of Lhcgr, Fshr, Inhbb, Ar, Esr1, Esr2, Gper and Cyp19a1 relative
to the expression of Rpl19. Two and a half micrograms of total Conflicts of interest
RNA were reverse transcribed using oligo(dT) primer with the There are no conflicts to declare.
help of a GoScript Reverse Transcription System kit (Promega,
Madison, USA) according to the manufacturer’s instructions.
Real-time PCR was carried out with an Applied Biosystems Acknowledgements
StepOnePlus Real-time PCR System (Applied Biosystems,
Singapore) using a SYBR Green qPCR Platinum SuperMix-UDG This study was supported by the Coordenação de
with the ROX (Life Technologies, Carlsbad, USA) kit under the Aperfeiçoamento de Pessoal de Nivel Superior, Brazil

112 | Toxicol. Res., 2018, 7, 102–116 This journal is © The Royal Society of Chemistry 2018
View Article Online

Toxicology Research Paper

[23038.009865/2013-32] and Fundacao Araucaria, Brazil [516/ 14 A. Mackevica, M. E. Olsson and S. F. Hansen, The release
2014]. of silver nanoparticles from commercial toothbrushes,
J. Hazard. Mater., 2017, 322(Part A), 270–275.
15 M. E. Quadros, R. Pierson, N. S. Tulve, R. Willis, K. Rogers,
Notes and references T. A. Thomas and L. C. Marr, Release of Silver from
Nanotechnology-Based Consumer Products for Children,
1 A. Ingle, A. Gade, S. Pierrat, C. Sonnichsen and M. Rai, Environ. Sci. Technol., 2013, 47, 8894–8901.
Mycosynthesis of silver nanoparticles using the fungus 16 OECD, Series on the Safety of Manufactured Nanomaterials,
Fusarium acuminatum and its activity against some 74, Exposure assessment of nano-silver (AgNP): case study,
human pathogenic bacteria, Curr. Nanosci., 2008, 4, 141– 2016, p. 36.
144. 17 S. C. o. E. a. N. I. H. R. SCENIHR, Nanosilver: safety, health
2 J. S. Kim, E. Kuk, K. N. Yu, J. H. Kim, S. J. Park, H. J. Lee, and environmental effects and role in antimicrobial resistance,
S. H. Kim, Y. K. Park, Y. H. Park, C. Y. Hwang, Y. K. Kim, 2014, p. 103.
Y. S. Lee, D. H. Jeong and M. H. Cho, Antimicrobial effects 18 R. M. Silva, D. S. Anderson, J. Peake, P. C. Edwards,
Published on 23 November 2017. Downloaded on 03/05/2018 03:38:16.

of silver nanoparticles, Nanomedicine, 2007, 3, 95–101. E. S. Patchin, T. Guo, T. Gordon, L. C. Chen, X. Sun,
3 J. Chen, C. M. Han, X. W. Lin, Z. J. Tang and S. J. Su, [Effect L. S. Van Winkle and K. E. Pinkerton, Aerosolized Silver
of silver nanoparticle dressing on second degree burn Nanoparticles in the Rat Lung and Pulmonary Responses
wound], Zhonghua waike zazhi, 2006, 44, 50–52. over Time, Toxicol. Pathol., 2016, 44, 673–686.
4 M. S. Cohen, J. M. Stern, A. J. Vanni, R. S. Kelley, 19 M. Almansour, Q. Jarrar, A. Battah and B. Jarrar,
E. Baumgart, D. Field, J. A. Libertino and Morphometric Alterations Induced by the Toxicity of
I. C. Summerhayes, In vitro analysis of a nanocrystalline Variable Sizes of Silver Nanoparticles, Int. J. Morphol., 2015,
silver-coated surgical mesh, Surg. Infect., 2007, 8, 397–403. 33, 544–552.
5 A. B. Lansdown, Silver in health care: antimicrobial effects 20 E. J. Park, J. Yi, Y. Kim, K. Choi and K. Park, Silver nano-
and safety in use, Curr. Probl. Dermatol., 2006, 33, 17–34. particles induce cytotoxicity by a Trojan-horse type mecha-
6 H. Y. Lee, H. K. Park, Y. M. Lee, K. Kim and S. B. Park, nism, Toxicol. in Vitro, 2010, 24, 872–878.
A practical procedure for producing silver nanocoated 21 F. T. Mathias, R. M. Romano, M. M. Kizys, T. Kasamatsu,
fabric and its antibacterial evaluation for biomedical appli- G. Giannocco, M. I. Chiamolera, M. R. Dias-da-Silva and
cations, Chem. Commun., 2007, 2959–2961, DOI: 10.1039/ M. A. Romano, Daily exposure to silver nanoparticles during
b703034g. prepubertal development decreases adult sperm and repro-
7 A. Dhawan and V. Sharma, Toxicity assessment of nano- ductive parameters, Nanotoxicology, 2015, 9, 64–70.
materials: methods and challenges, Anal. Bioanal. Chem., 22 H. K. Sleiman, R. M. Romano, C. A. Oliveira and
2010, 398, 589–605. M. A. Romano, Effects of prepubertal exposure to silver
8 N. Durán, P. D. Marcato, R. D. Conti, O. L. Alves, nanoparticles on reproductive parameters in adult male
F. T. M. Costa and M. Brocchi, Potential use of silver nano- Wistar rats, J. Toxicol. Environ. Health, Part A, 2013, 76,
particles on pathogenic bacteria, their toxicity and possible 1023–1032.
mechanisms of action, J. Braz. Chem. Soc., 2010, 21, 949– 23 S. A. Bidgoli, M. Mahdavi, S. M. Rezayat, M. Korani,
959. A. Amani and P. Ziarati, Toxicity assessment of nanosilver
9 R. Garcia-Contreras, L. Argueta-Figueroa, C. Mejia- wound dressing in Wistar rat, Acta Med. Iran., 2013, 51,
Rubalcava, R. Jimenez-Martinez, S. Cuevas-Guajardo, 203–208.
P. A. Sanchez-Reyna and H. Mendieta-Zeron, Perspectives 24 M. Korani, S. M. Rezayat and S. Arbabi Bidgoli, Sub-chronic
for the use of silver nanoparticles in dental practice, Int. Dermal Toxicity of Silver Nanoparticles in Guinea Pig:
Dent. J., 2011, 61, 297–301. Special Emphasis to Heart, Bone and Kidney Toxicities,
10 M. Horie, H. Kato, K. Fujita, S. Endoh and H. Iwahashi, In Iran. J. Pharm. Res., 2013, 12, 511–519.
vitro evaluation of cellular response induced by manufac- 25 C. Recordati, M. De Maglie, S. Bianchessi, S. Argentiere,
tured nanoparticles, Chem. Res. Toxicol., 2012, 25, 605–619. C. Cella, S. Mattiello, F. Cubadda, F. Aureli, M. D’Amato,
11 N. R. Savithramma, M. L. Rao, K. Rukmini and P. S. Devi, A. Raggi, C. Lenardi, P. Milani and E. Scanziani, Tissue dis-
Antimicrobial activity of silver nanoparticles synthesized by tribution and acute toxicity of silver after single intravenous
using medicinal plants, Int. J. ChemTech Res., 2011, 3, 9. administration in mice: nano-specific and size-dependent
12 W. W. I. C. f. S. WWICS, Project on Emerging effects, Part. Fibre Toxicol., 2016, 13, 12.
Nanotechnologies: Nanotechnology, http://www.nanotech- 26 H. Guo, J. Zhang, M. Boudreau, J. Meng, J. J. Yin, J. Liu and
project.org/, (accessed 09/06/2016, 2016). H. Xu, Intravenous administration of silver nanoparticles
13 E. McGillicuddy, I. Murray, S. Kavanagh, L. Morrison, causes organ toxicity through intracellular ROS-related loss
A. Fogarty, M. Cormican, P. Dockery, M. Prendergast, of inter-endothelial junction, Part. Fibre Toxicol., 2016, 13,
N. Rowan and D. Morris, Silver nanoparticles in the 21.
environment: Sources, detection and ecotoxicology, Sci. 27 C. Carlson, S. M. Hussain, A. M. Schrand, L. K. Braydich-
Total Environ., 2017, 575, 231–246. Stolle, K. L. Hess, R. L. Jones and J. J. Schlager, Unique cel-

This journal is © The Royal Society of Chemistry 2018 Toxicol. Res., 2018, 7, 102–116 | 113
View Article Online

Paper Toxicology Research

lular interaction of silver nanoparticles: size-dependent in regulation of hypothalamus-pituitary-testis axis and


generation of reactive oxygen species, J. Phys. Chem. B, spermatogenesis, Front. Endocrinol., 2014, 5, 1.
2008, 112, 13608–13619. 42 R. J. Aitken, Sperm function tests and fertility,
28 N. Hadrup, K. Loeschner, A. Mortensen, A. K. Sharma, Int. J. Androl., 2006, 29, 69–75; discussion 105–108.
K. Qvortrup, E. H. Larsen and H. R. Lam, The similar 43 H. Rodriguez-Martinez, Laboratory semen assessment and
neurotoxic effects of nanoparticulate and ionic silver in prediction of fertility: still utopia?, Reprod. Domest. Anim.,
vivo and in vitro, Neurotoxicology, 2012, 33, 416–423. 2003, 38, 312–318.
29 N. Asare, C. Instanes, W. J. Sandberg, M. Refsnes, 44 T. Rijsselaere, A. Van Soom, S. Tanghe, M. Coryn, D. Maes
P. Schwarze, M. Kruszewski and G. Brunborg, Cytotoxic and A. de Kruif, New techniques for the assessment of
and genotoxic effects of silver nanoparticles in testicular canine semen quality: a review, Theriogenology, 2005, 64,
cells, Toxicology, 2012, 291, 65–72. 706–719.
30 P. V. AshaRani, G. Low Kah Mun, M. P. Hande and 45 B. Lucas, C. Fields and M. C. Hofmann, Signaling pathways
S. Valiyaveettil, Cytotoxicity and Genotoxicity of Silver in spermatogonial stem cells and their disruption by toxi-
Nanoparticles in Human Cells, ACS Nano, 2009, 3, 279–290. cants, Birth Defects Res., Part C, 2009, 87, 35–42.
Published on 23 November 2017. Downloaded on 03/05/2018 03:38:16.

31 L. Xu, A. Shao, Y. Zhao, Z. Wang, C. Zhang, Y. Sun, J. Deng 46 H. Kato, M. Suzuki, K. Fujita, M. Horie, S. Endoh,
and L. L. Chou, Neurotoxicity of Silver Nanoparticles in Rat Y. Yoshida, H. Iwahashi, K. Takahashi, A. Nakamura and
Brain After Intragastric Exposure, J. Nanosci. Nanotechnol., S. Kinugasa, Reliable size determination of nanoparticles
2015, 15, 4215–4223. using dynamic light scattering method for in vitro toxi-
32 T. T. Win-Shwe and H. Fujimaki, Nanoparticles and neuro- cology assessment, Toxicol. in Vitro, 2009, 23, 927–934.
toxicity, Int. J. Mol. Sci., 2011, 12, 6267–6280. 47 L. Pinďáková, V. Kašpárková, K. Kejlová, M. Dvořáková,
33 L. Braydich-Stolle, S. Hussain, J. J. Schlager and D. Krsek, D. Jírová and L. Kašparová, Behaviour of silver
M. C. Hofmann, In vitro cytotoxicity of nanoparticles in nanoparticles in simulated saliva and gastrointestinal
mammalian germline stem cells, Toxicol. Sci., 2005, 88, fluids, Int. J. Pharm., 2017, 527, 12–20.
412–419. 48 M. van der Zande, R. J. Vandebriel, E. Van Doren,
34 L. K. Braydich-Stolle, B. Lucas, A. Schrand, R. C. Murdock, E. Kramer, Z. Herrera Rivera, C. S. Serrano-Rojero,
T. Lee, J. J. Schlager, S. M. Hussain and M. C. Hofmann, E. R. Gremmer, J. Mast, R. J. B. Peters, P. C. H. Hollman,
Silver nanoparticles disrupt GDNF/Fyn kinase signaling in P. J. M. Hendriksen, H. J. P. Marvin,
spermatogonial stem cells, Toxicol. Sci., 2010, 116, 577–589. Ad A. C. M. Peijnenburg and H. Bouwmeester,
35 C. Castellini, S. Ruggeri, S. Mattioli, G. Bernardini, Distribution, Elimination, and Toxicity of Silver
L. Macchioni, E. Moretti and G. Collodel, Long-term effects Nanoparticles and Silver Ions in Rats after 28-Day Oral
of silver nanoparticles on reproductive activity of rabbit Exposure, ACS Nano, 2012, 6, 7427–7442.
buck, Syst. Biol. Reprod. Med., 2014, 60, 143–150. 49 G. Bachler, N. von Goetz and K. Hungerbühler, A physio-
36 U. B. Kaiser, A. Jakubowiak, A. Steinberger and W. W. Chin, logically based pharmacokinetic model for ionic silver and
Differential Effects of Gonadotropin-Releasing Hormone silver nanoparticles, Int. J. Nanomed., 2013, 8, 3365–
(GnRH) Pulse Frequency on Gonadotropin Subunit and 3382.
GnRH Receptor Messenger Ribonucleic Acid Levels in Vitro 50 Y. S. Kim, J. S. Kim, H. S. Cho, D. S. Rha, J. M. Kim,
1, Endocrinology, 1997, 138, 1224–1231. J. D. Park, B. S. Choi, R. Lim, H. K. Chang, Y. H. Chung,
37 L. Nagirnaja, K. Rull, L. Uusküla, P. Hallast, M. Grigorova I. H. Kwon, J. Jeong, B. S. Han and I. J. Yu, Twenty-eight-
and M. Laan, Genomics and genetics of gonadotropin day oral toxicity, genotoxicity, and gender-related tissue dis-
beta-subunit genes: unique FSHB and duplicated LHB/CGB tribution of silver nanoparticles in Sprague-Dawley rats,
loci, Mol. Cell. Endocrinol., 2010, 329, 4–16. Inhalation Toxicol., 2008, 20, 575–583.
38 K.-H. Jeong and U. B. Kaiser, in Knobil and Neill’s 51 A. P. Abreu and U. B. Kaiser, Pubertal development and
Physiology of Reproduction, ed. J. D. Neill, T. M. Plant, regulation, Lancet Diabetes Endocrinol., 2016, 4, 254–
D. W. Pfaff, J. R. G. Challis, D. M. de Kretser, J. S. Richards 264.
and P. M. Wassarman, Academic Press, St Louis, 3rd edn, 52 S. R. Ojeda and M. K. Skinner, in Knobil and Neill’s
2006, vol. 1, pp. 1635–1701. Physiology of Reproduction, ed. T. M. Plant, D. W. Pfaff,
39 A. E. O’Connor and D. M. De Kretser, Inhibins in normal J. R. G. Challis, D. M. d. Kretser, J. S. Richards and
male physiology, Semin. Reprod. Med., 2004, 22, 177–185. P. M. Wassarman, Academic Press, St Louis, 3rd edn, 2006,
40 L. O’Donnell, S. J. Meachem, P. G. Stanton and pp. 2061–2126. DOI: 10.1016/B978-012515400-0/50043-9.
R. I. McLachlan, in Knobil and Neill’s Physiology of 53 D. Nesan and D. M. Kurrasch, Genetic programs of the
Reproduction, ed. J. D. Neill, T. M. Plant, D. W. Pfaff, developing tuberal hypothalamus and potential mecha-
J. R. G. Challis, D. M. de Kretser, J. S. Richards and nisms of their disruption by environmental factors, Mol.
P. M. Wassarman, Academic Press, St Louis, 3rd edn, 2006, Cell. Endocrinol., 2016, 438, 3–17.
vol. 1, pp. 1017–1069. 54 C. Korenbrot, I. Huhtaniemi and R. Weiner, Preputial sep-
41 A. Chimento, R. Sirianni, I. Casaburi and V. Pezzi, Role of aration as an external sign of pubertal development in the
estrogen receptors and g protein-coupled estrogen receptor male rat, Biol. Reprod., 1977, 17, 298–303.

114 | Toxicol. Res., 2018, 7, 102–116 This journal is © The Royal Society of Chemistry 2018
View Article Online

Toxicology Research Paper

55 R. S. Ge, G. R. Chen, Q. Dong, B. Akingbemi, C. M. Sottas, role of estrogens in male gonad : a review, Reprod. Biol.
M. Santos, S. C. Sealfon, D. J. Bernard and M. P. Hardy, Endocrinol., 2003, 1, 35.
Biphasic effects of postnatal exposure to diethyl- 67 L. O’Donnell, S. J. Meachem, P. G. Stanton and
hexylphthalate on the timing of puberty in male rats, R. I. McLachlan, in Knobil and Neill’s Physiology of
J. Androl., 2007, 28, 513–520. Reproduction, ed. J. D. Neill, T. M. Plant, D. W. Pfaff,
56 T. E. Stoker, L. G. Parks, L. E. Gray and R. L. Cooper, J. R. G. Challis, D. M. d. Kretser, J. S. Richards and
Endocrine-disrupting chemicals: prepubertal exposures P. M. Wassarman, Academic Press, St Louis, 3rd edn, 2006,
and effects on sexual maturation and thyroid function in pp. 1017–1069, DOI: 10.1016/B978-012515400-0/50026-9.
the male rat. A focus on the EDSTAC recommendations. 68 R. A. Hess, Estrogen in the adult male reproductive tract:
Endocrine Disrupter Screening and Testing Advisory A review, Reprod. Biol. Endocrinol., 2003, 1, 1–14.
Committee, Crit. Rev. Toxicol., 2000, 30, 197–252. 69 E. M. Eddy, T. F. Washburn, D. O. Bunch, E. H. Goulding,
57 Y. S. Kim, M. Y. Song, J. D. Park, K. S. Song, H. R. Ryu, B. C. Gladen, D. B. Lubahn and K. S. Korach, Targeted dis-
Y. H. Chung, H. K. Chang, J. H. Lee, K. H. Oh and ruption of the estrogen receptor gene in male mice causes
B. J. Kelman, Subchronic oral toxicity of silver nano- alteration of spermatogenesis and infertility, Endocrinology,
Published on 23 November 2017. Downloaded on 03/05/2018 03:38:16.

particles, Part. Fibre Toxicol., 2010, 7, 1. 1996, 137, 4796–4805.


58 H. Kanasaki, A. Oride, T. Mijiddorj, U. Sukhbaatar and 70 T. F. Lucas, E. R. Siu, C. A. Esteves, H. P. Monteiro,
S. Kyo, How is GnRH regulated in GnRH-producing C. A. Oliveira, C. S. Porto and M. F. Lazari, 17beta-estradiol
neurons? Studies using GT1-7 cells as a GnRH-producing induces the translocation of the estrogen receptors ESR1
cell model, Gen. Comp. Endocrinol., 2017, 247, 138–142. and ESR2 to the cell membrane, MAPK3/1 phosphorylation
59 L. Hu, R. L. Gustofson, H. Feng, P. Ki Leung, N. Mores, and proliferation of cultured immature rat Sertoli cells,
L. Z. Krsmanovic and K. J. Catt, Converse Regulatory Biol. Reprod., 2008, 78, 101–114.
Functions of Estrogen Receptor-α and -β Subtypes 71 T. F. Lucas, C. Royer, E. R. Siu, M. F. Lazari and C. S. Porto,
Expressed in Hypothalamic Gonadotropin-Releasing Expression and signaling of G protein-coupled estrogen
Hormone Neurons, Mol. Endocrinol., 2008, 22, 2250–2259. receptor 1 (GPER) in rat sertoli cells, Biol. Reprod., 2010, 83,
60 J. L. Temple, E. Laing, A. Sunder and S. Wray, Direct action 307–317.
of estradiol on gonadotropin-releasing hormone-1 neuro- 72 T. F. Lucas, M. F. Lazari and C. S. Porto, Differential role of
nal activity via a transcription-dependent mechanism, the estrogen receptors ESR1 and ESR2 on the regulation of
J. Neurosci., 2004, 24, 6326–6333. proteins involved with proliferation and differentiation of
61 J. Lindzey, W. C. Wetsel, J. F. Couse, T. Stoker, R. Cooper Sertoli cells from 15-day-old rats, Mol. Cell. Endocrinol.,
and K. S. Korach, Effects of castration and chronic steroid 2014, 382, 84–96.
treatments on hypothalamic gonadotropin-releasing 73 G. Cacciola, T. Chioccarelli, S. Fasano, R. Pierantoni and
hormone content and pituitary gonadotropins in male G. Cobellis, Estrogens and Spermiogenesis: New Insights
wild-type and estrogen receptor-alpha knockout mice, from Type 1 Cannabinoid Receptor Knockout Mice,
Endocrinology, 1998, 139, 4092–4101. Int. J. Endocrinol., 2013, 2013, 12.
62 O. S. Rissland, The organization and regulation of mRNA- 74 B. Robaire, B. T. Hinton and M.-C. Orgebin-Crist, in Knobil
protein complexes, Wiley Interdiscip. Rev.: RNA, 2017, 8, and Neill’s Physiology of Reproduction, ed. D. N. Jimmy,
e1369. M. P. Tony, W. P. Donald, R. G. C. John, M. d. K. David,
63 P. Bargi-Souza, R. M. Romano, F. Goulart-Silva, S. R. JoAnne and M. Wassarman, Academic Press, St Louis,
E. L. Brunetto and M. T. Nunes, T(3) rapidly regulates 3rd edn, 2006, pp. 1071–1148, DOI: 10.1016/B978-
several steps of alpha subunit glycoprotein (CGA) synthesis 012515400-0/50027-0.
and secretion in the pituitary of male rats: Potential reper- 75 D. D. Ricker, The autonomic innervation of the epididymis:
cussions on TSH, FSH and LH secretion, Mol. Cell. its effects on epididymal function and fertility, J. Androl.,
Endocrinol., 2015, 409, 73–81. 1998, 19, 1–4.
64 K. De Gendt, J. V. Swinnen, P. T. Saunders, L. Schoonjans, 76 H. Woelders, Overview of in vitro methods for evaluation of
M. Dewerchin, A. Devos, K. Tan, N. Atanassova, semen quality, Reprod. Domest. Anim., 1991, 1, 145–
F. Claessens and C. Lécureuil, A Sertoli cell-selective knock- 164.
out of the androgen receptor causes spermatogenic arrest 77 A. Amaral, B. Lourenco, M. Marques and J. Ramalho-
in meiosis, Proc. Natl. Acad. Sci. U. S. A., 2004, 101, 1327– Santos, Mitochondria functionality and sperm quality,
1332. Reproduction, 2013, 146, R163–R174.
65 R.-S. Wang, S. Yeh, L.-M. Chen, H.-Y. Lin, C. Zhang, J. Ni, 78 C. Pang, D. Hristozov, A. Zabeo, L. Pizzol, M. P. Tsang,
C.-C. Wu, P. A. di Sant’Agnese, K. L. deMesy-Bentley and P. Sayre and A. Marcomini, Probabilistic approach for
C.-R. Tzeng, Androgen receptor in Sertoli cell is essential assessing infants’ health risks due to ingestion of nano-
for germ cell nursery and junctional complex formation in scale silver released from consumer products, Environ. Int.,
mouse testes, Endocrinology, 2006, 147, 5624–5633. 2017, 99, 199–207.
66 S. Carreau, S. Lambard, C. Delalande, I. Denis-Galeraud, 79 USA, United States Pharmacopoeia. The official compendia of
B. Bilinska and S. Bourguiba, Aromatase expression and standard, Arabswell, New York, USA, 30 edn, 2012.

This journal is © The Royal Society of Chemistry 2018 Toxicol. Res., 2018, 7, 102–116 | 115
View Article Online

Paper Toxicology Research

80 R. M. Romano, S. N. Gomes, N. C. Cardoso, L. Schiessl, 82 G. Robb, R. Amann and G. Killian, Daily sperm production
M. A. Romano and C. A. Oliveira, New insights for male and epididymal sperm reserves of pubertal and adult rats,
infertility revealed by alterations in spermatic function and J. Reprod. Fertil., 1978, 54, 103–107.
differential testicular expression of thyroid-related genes, 83 P. Chomczynski and N. Sacchi, Single-step method of RNA
Endocrine, 2017, 55, 607–617. isolation by acid guanidinium thiocyanate-phenol-
81 M. Romano, R. Romano, L. Santos, P. Wisniewski, chloroform extraction, Anal. Biochem., 1987, 162, 156–
D. Campos, P. Souza, P. Viau, M. Bernardi, M. Nunes and 159.
C. Oliveira, Glyphosate impairs male offspring reproductive 84 K. J. Livak and T. D. Schmittgen, Analysis of relative gene
development by disrupting gonadotropin expression, Arch. expression data using real-time quantitative PCR and the 2
Toxicol., 2012, 86, 663–673. (-Delta Delta C(T)) Method, Methods, 2001, 25, 402–408.
Published on 23 November 2017. Downloaded on 03/05/2018 03:38:16.

116 | Toxicol. Res., 2018, 7, 102–116 This journal is © The Royal Society of Chemistry 2018

Вам также может понравиться