You are on page 1of 16

Nanotoxicology

ISSN: 1743-5390 (Print) 1743-5404 (Online) Journal homepage: https://www.tandfonline.com/loi/inan20

Acute reproductive toxicology after intratesticular


injection of silver nanoparticles (AgNPs) in Wistar
rats

Juliana Lis Mendes de Brito, Vanessa Nicolau de Lima, Dorleta Otaegui Ansa,
Sergio Enrique Moya, Paulo Cesar Morais, Ricardo Bentes de Azevedo &
Carolina Madeira Lucci

To cite this article: Juliana Lis Mendes de Brito, Vanessa Nicolau de Lima, Dorleta Otaegui Ansa,
Sergio Enrique Moya, Paulo Cesar Morais, Ricardo Bentes de Azevedo & Carolina Madeira Lucci
(2020): Acute reproductive toxicology after intratesticular injection of silver nanoparticles (AgNPs) in
Wistar rats, Nanotoxicology, DOI: 10.1080/17435390.2020.1774812

To link to this article: https://doi.org/10.1080/17435390.2020.1774812

View supplementary material

Published online: 12 Jun 2020.

Submit your article to this journal

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=inan20
NANOTOXICOLOGY
https://doi.org/10.1080/17435390.2020.1774812

ARTICLE

Acute reproductive toxicology after intratesticular injection of silver


nanoparticles (AgNPs) in Wistar rats
Juliana Lis Mendes de Britoa , Vanessa Nicolau de Limaa , Dorleta Otaegui Ansab , Sergio Enrique
Moyac , Paulo Cesar Moraisd,e , Ricardo Bentes de Azevedof and Carolina Madeira Luccia
a
Laboratory of Animal Reproduction, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasilia,
Brasilia, Brazil; bMass Spectrometry Platform, Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and
Technology Alliance (BRTA), San Sebastian, Spain; cSoft Matter Nanotechnology Lab, Center for Cooperative Research in Biomaterials
(CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), San Sebastian, Spain; dInstitute of Physics, University of Brasilia,
Brasilia, Brazil; eGenomic Sciences and Biotechnology, Catholic University of Brasilia, Brasilia, Brazil; fLaboratory of Nanotechnology,
Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasilia, Brasilia, Brazil

ABSTRACT ARTICLE HISTORY


This study aimed to evaluate the effects of an intratesticular injection of silver nanoparticles Received 28 February 2020
(AgNPs) on reproductive parameters and health of rats, and to evaluate the AgNPs biodistribu- Revised 22 April 2020
tion in order to develop a nanotechnological contraceptive agent for male animals. Treated ani- Accepted 23 May 2020
mals received 220 lL of AgNPs solution (0.46 mg-Ag/ml) in each testicle and were euthanized:
KEYWORDS
seven, 14, 28, and 56 days after injection. A significant decrease (p < 0.05) in the percentage of Reproductive toxicity; sperm
motile sperm in D7 (8.8%) was observed, comparing to the control (73.3%), D14 (86.0%), D28 morphology; sperm motility;
(68.2%), and D56 (90.0%) groups. D7 group also presented a decrease (p < 0.05) in the percent- seminiferous tubules;
age of normal spermatozoa. Additionally, D7 group showed an increase (p < 0.05) in abnormal nanosilver; rats
midpiece and sperm head morphology compared to the Control group. Seminiferous tubules
presented all germline cell types and spermatozoa for all groups. However, D7 group did not
present spermatozoa in the epididymis, whereas some spermatozoa and cellular debris were vis-
ible in D14 and D28 groups. All animals presented hematological parameters, creatinine, and
alanine aminotransferase values within the normal limits for Wistar rats. The percentage of silver
found in the liver was always higher than in the other organs analyzed. A pioneering mathemat-
ical model is proposed, from which the half-life time of silver in the liver (17 days), spleen
(23 days), lungs (30 days), and kidneys (35 days) was extracted. In conclusion, some acute and
severe toxic effects were observed in sperm cells following intratesticular injection of AgNPs,
although these effects were reversible. No adverse effects to general animal health were
observed.

Introduction Amaku, Dias, and Ferreira 2010) particularly because


It is well documented that many nanoparticles (NPs) stray animals can transmit diseases to domestic ani-
are capable of crossing the hemato-testicular barrier, mals and humans (Slater 2002; Slater and Shain
leading to toxic effects on male reproductive func- 2005; FAO 2011), not to mention preying on wild
tions such as reducing sperm count and altering animals. In the same way, exotic invasive animals,
sperm cell function and cell viability (Braydich-Stolle as cats in Australia, rabbits in New Zealand, and
et al. 2010; Ema et al. 2010; Kim et al. 2010). feral pigs in Brazil, cause ecological and economic
Profiting from advances in the nanobiotechnology problems. The major concern regarding these ani-
field, it is possible to make use of the already known mal populations is their high reproductive potential
toxicity of some nanomaterials to develop alternative (Olson and Johnston 1993). It is well established
methods to neuter stray animals. that the most effective way to control and reduce
The overpopulation of stray animals (cats and the number of street animals is their sterilization
dogs) is a major public health problem (Slater 2001; (Jana and Samanta 2007). Nevertheless, surgical

CONTACT Carolina Madeira Lucci carollucci@gmail.com Laboratory of Animal Reproduction, Department of Physiological Sciences, Institute of
Biological Sciences, Campus Universitario Darcy Ribeiro, Brasilia, DF 70910-900 Brazil
Supplemental data for this article can be accessed here
ß 2020 Informa UK Limited, trading as Taylor & Francis Group
2 J. L. M. DE BRITO ET AL.

castration programs currently available are not suffi- Given the toxic effects of AgNPs in the repro-
ciently effective on big populations. This way, there ductive system following systemic injection, we
is a huge need for alternative methods for castra- decided to explore the potential of AgNPs as a ster-
tion/contraception for animals. ilizing agent, maximizing its activity by a local intra-
Since the 1960 researchers are looking for simple testicular injection. This approach could be used as
methods to neuter male animals, requiring for a strategy to reduce fertility/reproduction in stray
example a single injection of a neutering drug. animals. Therefore, the aims of this study were to (i)
Many attempts to find effective drugs to produce evaluate the effects of an AgNP intratesticular injec-
the so called chemical castration have been carried tion on reproductive parameters and health and (ii)
out using calcium chloride (Jana and Samanta 2006, evaluate the AgNP biodistribution after intratesticu-
2007, 2011; Leoci et al. 2019), cadmium chloride lar administration.
(Kar 1961), ferric chloride and ferrous sulfate (Kar
et al. 1965), glycerol (Wiebe et al. 1989), lactic acid Material and methods
(Fordyce et al. 1989), and zinc gluconate by intra-
testicular injection. In fact, the intratesticular treat- Silver nanoparticle preparation and
ment with an injectable zinc gluconate (neutralized characterization
in arginine) has been approved in 2003 by the Silver nanopowder containing polyvinylpyrrolidone
Food and Drug Administration (FDA) for use in (PVP) as dispersant was obtained from Sigma-
dogs (ACC&D 2016). However, it has not been Aldrich (St. Louis, MO, CAS number 7440-22-4).
widely used due to side effects and researches are According to the AgNPs Safety Data Sheet provided
still being performed. We saw in the nanotechnol- by Sigma-Aldrich, the particle size was <100 nm. An
ogy a potential tool to develop an alternative cas- AgNPs suspension was prepared according to
tration/contraception method, an aspect that only a Caceres-Velez et al. (2018). Briefly, the powder was
few research groups have addressed until now (Liu diluted in ultrapure water at a 0.46 mg-Ag/ml con-
et al. 2015a, 2015b; Yostawonkul et al. 2017). centration. The suspension was sonicated (Vibra-
Silver nanoparticles (AgNPs) are known to induce CellTM 75042, 20 kHz, 750 W) on ice for 40 min, with
toxic effects on spermatogenesis. Studies have pulsing for 5/3 (ON/OFF) seconds and with the
reported that AgNPs cause: DNA damage in germ amplitude of 20%.
cells, decrease sperm count and promote morpho- The stock solution (pH 7.49) was diluted (1:4) in
metric changes in seminiferous tubules ultrapure water while particle size and surface
(Dziendzikowska et al. 2012; Gromadzka-Ostrowska charge were measured in triplicate with 20 sub-runs
et al. 2012); decrease spermatogonia proliferation by dynamic light scattering (DSL), using a Zetasizer
(Braydich-Stolle et al. 2005, 2010), and increase Nano-ZS90 (Malvern, Malvern Hills, UK). Samples of
germ cell apoptosis (Garcia et al. 2014). In an ele- this diluted solution were dripped onto a 300-mesh
gant study, Zhang et al. (2015) demonstrated that copper grid, dried overnight at room temperature
AgNPs induced massive cell death in TM3 (Leydig (RT), and analyzed in a transmission electron micro-
cell line) and TM4 (Sertoli cell line) cells, and inhib- scope (TEM) system (JEM-2100F, JEOL, Tokyo,
ited proliferation and self-renewal of spermatogon- Japan). Images were taken at 20 000x and 50 000x
ial stem cells (SSCs). In this study AgNPs induced magnification and the sizes of 1000 particles cap-
expression of autophagy-related genes and acti- tured. Other characteristics of AgNPs are described
vated signaling molecules involved in apoptosis. in Caceres-Velez et al. (2018).
The mRNA levels of proteins that play important
roles in self-renewal, proliferation, meiosis, and dif-
Experimental design
ferentiation of SSCs were also significantly lower in
AgNPs-treated cells, suggesting that AgNPs could Animal experimentation was approved by the Ethics
affect the development of germ cells in the testes. Committee on Animal Use of the University of
Finally, they proved that AgNPs toxicity in those Brasilia (CEUA-UnB - n 102854/2015). A total of 28
cells was due to reactive oxygen species (ROS) gen- male Wistar rats (8–9 weeks old and weighting
eration and accumulation. 154 ± 25 g) were kept in polypropylene cages with
NANOTOXICOLOGY 3

woodchips as bedding, in a ventilated room with parameters were evaluated: white blood cell count
temperature 24  C and under a 12 h/12h light/ (WBC), red blood cell count (RBC), hemoglobin con-
dark cycle with ad libitum access to commercial centration (HB), hematocrit (HTC), mean corpuscular
food (Nutrina - Average nutrient content/Kg: pro- volume (MCV), and mean corpuscular hemoglobin
tein-minimum 230 g, total fat- minimum 25 g, crude (MCH) using an automated hematology analyzer
fibre-maximum 90 g, crude ash-maximum 80 g, cal- (Sysmex pocH – 110 iV Diff TM, Japan). The bio-
cium-maximum 18 g, and phosphorus-minimum 8 g) chemical parameters evaluated were alanine amino-
and tap water. Initially, animals were randomly div- transferase (ALT) and aspartate aminotransferase
ided into three groups: (1) animals without any (AST) for liver function, and urea and creatinine for
manipulation (Control group: N ¼ 4), (2) animals kidney function using specific assay kits (Labmax
100 - LabtestV, Brazil).
R
receiving an intratesticular injection of saline solu-
tion (Sham group: N ¼ 4), and (3) animals receiving
an intratesticular injection of the AgNPs solution
Organs morphometrics
(AgNPs group: N ¼ 20). All animals from the Sham
and AgNPs groups were anesthetized with ketamine The testicles and epididymis were removed, sepa-
(90 mg/kg) and xylazine (10 mg/kg) (SyntecV R ) prior rated, measured, and weighed. The liver, spleen,
to intratesticular injection. Each animal from the lungs, and kidneys were also weighed. The absolute
AgNPs group received 220 lL of AgNPs solution in weight of each organ was transformed into relative
each testicle (corresponding to 100 lg Ag/testicle, weight (organ weight/body weight 100).
which is equivalent to 50 lg Ag/g testicular tissue), Testicular length and width measurements were
in two different spots. The chosen dose was based used to determine testicular volume (VOL). For this,
on in vitro studies that calculated the EC50 (50% the average of two mathematical formulae was used:
effective concentration) of AgNPs to produce toxic the cylinder (VOLC ¼ 2  [p( width/2)2(length)]) and
effects in male reproductive cells was 8.75 lg/ml the prolate spheroid (VOLP ¼ 2  [(4/3)p(width/
(Braydich-Stolle et al. 2005) and that concentrations 2)2(length/2)]), which together form the closest value
10 lg/ml, Ag-NPs induced a significant decline in to the true testicle volume (Louvandini et al. 2008).
SSC proliferation (Braydich-Stolle et al. 2010). To
maximize the effects in vivo we used 5 times the Testicles and epididymis processing
dose used in vitro. Animals from the Sham group
received 0.9% sterile saline solution in the same vol- The left epididymis was minced in 2 ml of warmed
ume and manner. Animals from the AgNPs group (37  C) saline solution to remove the spermatozoa.
were subdivided into four subgroups, according to One drop was immediately placed on a slide, cov-
the day of euthanasia (D7, D14, D28, and D56; ered with a cover slip and analyzed for sperm motil-
N ¼ 5 in each subgroup). Animals from the Control ity. In order to evaluate sperm morphology, 1 ml of
group were euthanized after 14 days, whereas ani- sperm suspension was added to 1 ml of 10% forma-
mals from the Sham group were euthanized seven - lin, with 100 sperm cells per sample classified
days after the start of the experiment. according to their morphology under a phase con-
The animals were weighed weekly and their trast microscope (Nikon Eclipse Ci, Tokyo, Japan).
behavior was observed daily for signs of pain and/or Both testicles and the right epididymis were fixed
inflammatory response. The animals were euthanized in Bouin’s fixative solution for at least 24 h, dehy-
by an anesthetic overdose (ketamine and xylazine) drated in a graded ethanol series, and embedded in
ParaplastV (Sigma Aldrich). Five nonconsecutive sec-
R

and cardiac puncture. Following euthanasia, blood,


both testicles and epididymis, liver, spleen, lungs, tions (5 mm thick) from each organ were mounted on
and kidneys were collected for further evaluation. slides, stained with hematoxylin and eosin, and eval-
uated under a light microscope (Nikon Eclipse Ci,).
Testicular sections were investigated for the pres-
Blood analysis
ence of spermatogenic lineage, Sertoli and Leydig
Blood was collected for hematological and bio- cells. Histomorphometric parameters were obtained
chemical analysis. The following hematological from 25 seminiferous tubule cross-sections for each
4 J. L. M. DE BRITO ET AL.

testicle. Tubule and tubular lumen diameter and tested for normality by the Shapiro-Wilk test. The var-
area, together with seminiferous epithelium iables were compared among groups using analysis
height, were measured using the ImageJ software of variance (ANOVA) and Tukey’s test. Differences
(National Institutes of Health, Bethesda, Maryland, were considered significant when p < 0.05.
Version 1.52a).
Results
Determination of silver concentration in organs AgNP characterization
One kidney and one half of each of the other The morphological characteristics of the AgNPs are
organs were frozen for silver quantification and bio- presented in Figure 1. According to the TEM micro-
distribution analyses. graphs, the AgNPs are approximately spherical, with a
Total silver concentration was measured by mean diameter of 23 ± 8 nm (varying from 3–46 nm).
inductively coupled plasma mass spectrometry (ICP- According to the DSL measurements, the mean
MS), with a two-step digestion protocol (Ashoka hydrodynamic diameter of the AgNPs is 215 nm
et al. 2009). Briefly, 2 ml of 70% nitric acid reacted (Polydispersity Index - PDI of 0.41), with a mean zeta
with the organs for three days at RT, under airflow. potential of 28.
All samples were diluted to reach a concentration
of 1.94%. Finally, 285 ml of each sample was diluted
Animal observation (body, epididymis, testicles,
in 10 ml of ultrapure water and promptly analyzed
and organ weight)
by ICP-MS (iCAPTM Q ICP-MS Thermo ScientificTM,
Waltham, MA) at the CIC biomaGUNE Mass facility In general, all animals exhibited normal weight gain
(San Sebastian, Spain). (Figure 2) and a healthy state throughout the study.
In addition, no sign of inflammatory process or pain
was observed.
Statistical analysis
The relative weight of testicles and epididymitis
Results were analyzed using the SPSS 22.0 (IBMV
R
showed no significant differences among groups
Statistics, Armonk, NY) package software. Data were (Table 1). The volume of the left testicle in the

Figure 1. Electron micrography of the AgNPs (upper left-hand side panel) and size distribution of the AgNPs by transmission elec-
tron microscope (TEM) measurement (upper right-hand side panel). Table (lower panel) presents the morphological parameters
(TEM and DSL) and zeta potential of the AgNPs.
NANOTOXICOLOGY 5

groups treated with AgNPs euthanized at D14, D28, Sperm motility and morphology
and D56 was significantly higher (p < 0.05) than in
A significant effect of AgNPs intratesticular injection
AgNP-treated group euthanized at D7 and the
was observed on sperm motility, with a significant
Sham group.
decrease (p < 0.05) in the percentage of motile
The general aspect of the liver, spleen, lungs,
sperm in the AgNPs-D7 group, in comparison to all
and kidneys was normal during euthanasia. No dif-
other groups, with the exception of the Sham
ferences in terms of aspect, color, or format were
group (Figure 3).
observed. The relative weight of the spleen
A significant decrease (p < 0.05) in the percent-
presents a higher value for the AgNPs-D28 group in
age of normal spermatozoa was also observed in
comparison with other groups, except in the Sham
the AgNPs-D7 group in comparison to all other
group. The relative weight of the liver also showed
groups (Figure 4). The AgNPs-D7 group also dem-
a higher value in the AgNPs-D7 group when com-
onstrated a significant increase (p < 0.05) in abnor-
pared with the other groups, apart from the Sham
mal morphology of the midpiece when compared
group (Table 2).
to all other groups, and a significant increase
(p < 0.05) in abnormal sperm head morphology
compared to the control group (Figure 4).

Hematological analysis
Mean (± standard deviation (SD)) values, together
with minimum and maximum values, for the hema-
tological analysis of each group are presented in
Table 3. Data from each animal was analyzed indi-
vidually. In general, all animals presented hemato-
logical parameters within the normal limits for
Wistar rats (last column in Table 3). In a few cases,
specific parameters were altered, but without correl-
ation with the treatments.
Mean (±SD) values, together with minimum and
maximum values, for the biochemical analysis for
Figure 2. Weekly weight gain in each study group during the each group are presented in Table 4, with the nor-
experimental period. mal limits for Wistar rats in the final column.

Table 1. Relative weight (%) of the testicles and epididymis, and testicular volume (cm3; mean ± standard deviation).
AgNPs- AgNPs- AgNPs- AgNPs-
Control group Sham group D7 D14 D28 D56
Right testicle 0.56 ± 0.01 0.58 ± 0.06 0.49 ± 0.17 0.56 ± 0.05 0.56 ± 0.05 0.54 ± 0.07
Left testicle 0.58 ± 0.03 0.57 ± 0.05 0.52 ± 0.15 0.56 ± 0.06 0.55 ± 0.04 0.49 ± 0.06
Right epididymides 0.26 ± 0.04 0.22 ± 0.06 0.31 ± 0.19 0.24 ± 0.06 0.24 ± 0.04 0.29 ± 0.07
Left epididymides 0.31 ± 0.16 0.30 ± 0.03 0.27 ± 0.04 0.22 ± 0.02 0.25 ± 0.03 0.29 ± 0.05
Right testicle volume (cm3) 3.13 ± 0.58 2.20 ± 0.67 2.40 ± 0.45 2.92 ± 0.31 2.52 ± 0.26 2.96 ± 0.34
Left testicle volume (cm3) 2.80 ± 0.23A,B 2.08 ± 0.24A 2.06 ± 0.63A 3.50 ± 0.47B 3.46 ± 0.53B 3.12 ± 0.50B
A,B
Indicate significant difference in the same line (p < 0.05).

Table 2. Relative weight (%) of organs (mean ± standard deviation).


AgNPs- AgNPs- AgNPs- AgNPs-
Control group Sham group D7 D14 D28 D56
Liver 4.70 ± 0.26A 4.87 ± 0.69A,B 5.96 ± 1.09B 4.56 ± 0.37A 4.39 ± 0.37A 3.69 ± 0.12A
Spleen 0.27 ± 0.04A 0.41 ± 0.13A,B 0.39 ± 0.06A 0.31 ± 0.03A 0.57 ± 0.05B 0.37 ± 0.12A
Lung 0.66 ± 0.09 0.63 ± 0.07 0.67 ± 0.23 0.70 ± 0.13 0.65 ± 0.06 0.59 ± 0.09
Kidney 0.40 ± 0.03 0.45 ± 0.08 0.46 ± 0.04 0.50 ± 0.17 0.44 ± 0.03 0.38 ± 0.02
A,B
Indicate significant difference in the same line (p < 0.05).
6 J. L. M. DE BRITO ET AL.

Individual analysis of each animal showed all ani-


mals presented creatinine and ALT values within
the normal limits for Wistar rats. However, all ani-
mals (treated and controls) presented urea values
higher than the normal limit for Wistar rats.
Concerning AST, all animals (treated and controls)
showed values higher than the normal limits.

Histomorphometry of seminiferous and


epidydimal tubules

Figure 3. Mean percentage of motile spermatozoa in each Histological analysis of the testicles allowed the
study group (A and B indicate significant difference, p < 0.05). identification of all germline cells in the seminifer-
ous tubules, as well as spermatozoa in the lumen of
the tubules, and normal architecture of the semin-
iferous tubule with the germinal epithelium at dif-
ferent stages of development for the animals of the
Control, Sham, and AgNPs-D56 groups. In the
AgNPs-D7, AgNPs-D14, and AgNPs-D28 groups, it
was possible to visualize that the architecture of
the seminiferous tubule is normal, despite the pres-
Figure 4. Mean percentage of normal and abnormal sperm
ence of some debris in the lumen and few sperma-
morphology by spermatic region (A and B indicate significant
difference in the same category, p < 0.05). tozoa (Figure 5).

Table 3. Hematological values in peripheral blood (mean ± standard deviation and minimum to maximum).
Normal limits
Parameter Control group Sham group AgNPs- D7 AgNPs- D14 AgNPs- D28 AgNPs- D56 for Wistar ratsa
WBC (103/mL) 4±2 4±5 4±2 6±4 4±1 5±2 6.0–15.0
(2.5–6.2) (0.8–12.5) (1.7–5.8) (1.2–9.7) (3.1–5.8) (2.4–7.4)
6
RBC (10 /mL) 5.5 ± 0.7 4±2 6±2 6±2 5.8 ± 0.9 7±1 6.0–10.0
(4.6–6.3) (1.2–6.8) (2.6–7.8) (3.5–7.3) (4.2–6.5) (5.3–8.3)
HB (g/dL) 12 ± 2 8±5 11 ± 4 13 ± 4 12 ± 2 15 ± 3 11.0–19.5
(9.6–13.2) (2.5–13.9) (5.0–15.3) (7.2–15.7) (8.5–13.6) (10.2–16.4)
HTC (%) 32 ± 4 26 ± 20 32 ± 10 35 ± 9 34 ± 6 40 ± 6 39–55
(26.3–36.9) (8.0–26.7) (14.5–44.9) (20.3–41.9) (24.6–38.0) (29.5–44.9)
MCV (fL) 58.1 ± 0.7 65 ± 3 58 ± 2 57.9 ± 0.6 59 ± 2 55 ± 1 46.0–65.0
(57.5–59.0) (60.8–67.1) (56.0–59.8) (57.1–58.5) (55.0–60.4) (53.1–55.7)
MCH (pg) 21.2 ± 0.7 20.5 ± 0.2 19.7 ± 0.4 21.1 ± 0.6 20.8 ± 0.5 19.7 ± 0.3 18.0–23.0
(20.7–22.3) (20.3–20.7) (19.3–20.4) (20.5–24.5) (20.2–21.6) (19.2–20.1)
MCHC (g/dL) 37 ± 1 31 ± 1 33.8 ± 0.7 37 ± 1 36.5 ± 0.9 36 ± 1 31.0–40.0
PLT (103/ mL) 267 ± 300 133 ± 200 299 ± 300 239 ± 90 330 ± 300 286 ± 200 200–1500
WBC: white blood cells; RBC: red blood cells; HB: hemoglobin; HTC: hematocrit; MCV: mean corpuscular volume; MCH: mean corpuscular hemoglobin.
As reported by Cubas, Silva, and Cat~ao-Dias (2014).
a

Table 4. Biochemical plasma variables (means ± standard deviation and minimum to maximum).
Normal Limits
Parameter Control group Sham group AgNPs- D7 AgNPs- D14 AgNPs- D28 AgNPs- D56 for Wistar ratsa
Creatinine (mg/dl) 0.70 ± 0.07 0.71 ± 0.05 0.59 ± 0.04 0.4 ± 0.2 0.45 ± 0.08 0.58 ± 0.04 0.2–0.8
(0.61–0.77) (0.66–0.78) (0.54–0.66) (0.20–0.63) (0.32–0.55) (0.53–0.61)
Urea (mg/dl) 65 ± 20 43 ± 3 64 ± 10 77 ± 8 82 ± 4 78 ± 4 15–21
(44–80) (43–49) (52–74) (64–84) (76–88) (73–84)
Alanine amino transferase (u/l) 55 ± 20 120 ± 60 55 ± 50 51 ± 20 48 ± 9 65 ± 20 17–224
(43–78) (58–178) (18–151) (29–71) (34–60) (39–89)
Aspartate aminotransferase (u/l) 186 ± 70 225 ± 100 170 ± 100 158 ± 50 129 ± 30 294 ± 70 39–92
(108–269) (115 –390) (93–313) (84–198) (100–169) (220 –393)
a
As reported by Cubas, Silva, and Cat~ao-Dias (2014).
NANOTOXICOLOGY 7

Figure 5. Micrographs of seminiferous tubules from each group. (a) Control group; (b) Sham group; (c) AgNPs-D7 group; (d)
AgNPs-D14 group; (e) AgNPs-D28 group; and (f) AgNPs-D56 group. (Bars ¼ 10 lm).

The histology of the epididymis showed Compared to the Control group, all tubular
spermatozoa in the lumen in the Control and parameters evaluated are significantly smaller
Sham groups, as well as in the AgNP-D56 (p < 0.05) in the AgNPs-D7 and Sham groups
group. In the AgNP-D7 group, it was not possible (Table 5). Furthermore, a significantly lower epithe-
to visualize any spermatozoa, whereas in the lium height in the AgNPs-D28 group and a signifi-
AgNPs-D14 and AgNPs-D28 groups some sperma- cant smaller lumen diameter in the AgNPs-D14
tozoa were visible together with some debris group were noted in comparison to the Control
(Figure 6). group values (p < 0.05).
8 J. L. M. DE BRITO ET AL.

Figure 6. Micrographs of epididymis from each group. (a) Control group; (b) Sham group; (c) AgNPs-D7 group; (d) AgNPs-D14
group; (e) AgNPs-D28 group; and (f) AgNPs-D56 group. (Bars ¼ 10 lm).

Table 5. Morphometry of seminiferous tubules (mean ± standard deviation) of all groups.


Control group Sham group AgNPs-D7 AgNPs-D14 AgNPs-D28 AgNPs-D56
Tubule area (mm2) 33166 ± 3700A,B 30514 ± 3000C 29213 ± 3000D 33144 ± 4000A,B 32549 ± 4000B 33979 ± 4000A
Lumen area (mm2) 14378 ± 4000A 12841 ± 3000B 11342 ± 3000C 13820 ± 4000A 14676 ± 3000A 14504 ± 4000A
Tubule diameter (mm) 276 ± 50A,B 250 ± 70C 247 ± 50C 273 ± 70A,B 269 ± 60B 280 ± 60A
Lumen diameter (mm) 106 ± 40A 97 ± 40B 75 ± 30C 97 ± 40B 105 ± 40A,B 104 ± 50A,B
Height of epithelium (mm) 90 ± 20A 82 ± 10C 86 ± 20C 94 ± 30B 84 ± 20C 93 ± 20A,B
A,B,C,D
Indicate significant difference in the same line (p < 0.05).

ICP-MS intratesticular injection, silver was found mainly in


The total amount of silver (C) in the organs (liver, the liver (0.82–2.64 mg-Ag), whereas the amount of
spleen, lungs, and kidneys) as a function of time (t) silver in the spleen (0.006–0.220 mg-Ag), lungs
was determined by ICP-MS (Figure 7). Following (0.10–0.87 mg-Ag) and kidneys (0.04–0.36 mg-Ag) was
NANOTOXICOLOGY 9

Figure 7. Mean amount of silver (lg-Ag) in the (a) liver, (b) spleen, (c) lungs, and (d) kidneys of animals treated with AgNPs ver-
sus time (t), in the range of seven to 56 days. Solid lines represent the best curve fitting to the experimental data (solid symbols).

the evaluated organs individually. In Figure 7, solid


lines represent the best curve fitting to the experi-
mental data (solid symbols) using the equation
described in the Supplementary Material. The math-
ematical model determined a lower half-life time in
the liver (17 days), followed by the spleen (23 days),
lungs (30 days), and kidneys (35 days). It is worth
mentioning that the mathematical model also sug-
gested that after the intratesticular injection of
Figure 8. Distribution of the total measured silver by the AgNPs, the NPs take longer to reach the analyzed
organs analyzed (liver, spleen, lungs, and kidneys) from ani- organs (liver, spleen, lungs, and kidneys). In the tes-
mals treated with AgNPs on days seven, 14, 28, and 56 after
intratesticular injection.
ticle, the mean amount of silver found decreased
over time (from 0.74 mg-Ag on D7 to 0.19 mg-Ag on
considerably lower. The data were fitted using a D56), but no significant differences were evidenced
mathematical model that enabled extraction of the (p > 0.05). The amount of silver found in the epididy-
half-life time (Th) of the injected AgNPs in each of mis was negligible (0.002–0.004 lg-Ag).
10 J. L. M. DE BRITO ET AL.

While injected into the testicles, the data ana- 14 days and returned back to normal 56 days after
lyzed allows us to conclude that there is a tendency injection. It has already been shown that AgNPs
of AgNPs to accumulate preferably in the liver, cause sperm damage. Ahmed, Abdelrahman, and
although the organ is capable of eliminating the Shalaby (2017) observed a significant reduction in
injected silver in a rather fast manner. sperm count and motility together with an increase
The percentage of silver found in each systemic in sperm abnormality in animals that received intra-
organ (liver, spleen, lungs, and kidneys) is shown in peritoneal injections of AgNPs (100 or 1000 mg/Kg)
Figure 8. The percentage of silver found in the liver daily for a total of seven or 28 days. Baki et al.
was significantly higher on D7 than D56 (p < 0.05). (2014) also observed a significant decrease in sperm
Conversely, the percentage of silver found in the motility and the number of sperm with normal
spleen was significantly higher (p < 0.05) on D56 morphology in Wistar rats that were fed AgNPs (25,
than on D7 (Figure 8), although the total silver 50, 100, or 200 mg/Kg) daily for a total of 45 days.
amount was lower (0.22 mg-Ag). Gromadzka-Ostrowska et al. (2012) observed a
decrease in the number of epididymal spermatozoa
and an increase in the number of dead sperm cells,
Discussion
together with significantly higher DNA damage in
For many years, researchers have looked for an alter- germ cells, after a single intravenous injection of
native neutering/contraceptive method for animals, AgNPs (5 or 10 mg/Kg). Although the doses and
especially stray dog and cats. A single intratesticular administration route greatly varies among studies,
injection that could cause the interruption of sperm- all of the aforementioned authors pointed out that
atogenesis would be particularly interesting for its sim- the nanotoxicity of AgNPs in sperm cells was
plicity and speed, as it could be applied to many enhanced with increasing dosage and nanoparticle
animals in a single day without the need for specific exposure (Gromadzka-Ostrowska et al. 2012; Baki
equipment or environment. In general, zinc gluconate et al. 2014; Ahmed, Abdelrahman, and Shalaby
and calcium chloride, the most used substances for 2017). In our study we used a relatively low dose
chemical castration by intratesticular injection, cause (equivalent to 1.25 mg/kg) compared with these
atrophy in the seminiferous tubules, disruption in previous studies; however, we used a local adminis-
spermatogenesis, fibrosis and calcification of testicular tration (50 lg/g of testicular tissue) instead of the
parenchyma and reduction of testicular volume, systemic administration used by the cited authors,
together with reduced sperm motility and low sperm where only a part (not known) of the dose would
count or azoospermia, leading to subfertility or infertil- reach the gonad. We envisaged that a local admin-
ity in male animals (Fagundes et al. 2014; Vannucchi istration would potentialize the toxic effects on the
et al. 2015; Silva et al. 2018; Leoci et al. 2019; spermatogenesis with minimal effects in systemic
Rafatmah, Mogheiseh, and Eshghi 2019). However, organs. Moreover, as these studies did not evaluate
many studies report side effects in the treated animals, the results over time, there is no mention of the
such as swelling, inflammation, pain, hemorrhage, effects being permanent or temporary.
necrosis and scrotal ulcerations (Levy et al. 2008; The local administration used in this work (intra-
Oliveira et al. 2013; Forzan et al. 2014; Rafatmah, testicular injection) was chosen to take advantage
Mogheiseh, and Eshghi 2019). In this sense, researches of a direct effect of the AgNPs on the sperm cell
are still needed to optimize these methods or find lineage. In an in vitro study with C18-4 cell line (a
new approaches to impair male animal reproduction. type A spermatogonia isolated from mouse testes)
This study intended to evaluate the effects of it was shown that AgNPs drastically reduced mito-
AgNPs injected directly in the testicles, with the chondrial function and cell viability, leading to
view to developing a contraceptive method for ani- necrosis and apoptosis of the cells (Braydich-Stolle
mals based on a single injection. We observed a et al. 2005). These toxic effect of AgNPs started
severe reduction in sperm motility and the number between 5 and 10 lg/ml, with an EC50 calculated
of sperm with normal morphology seven days after at 8.75 lg/ml (Braydich-Stolle et al. 2005). In the
the treatment, although these findings were not present study, the dose used was 50 lg/g of tes-
long lasting. Sperm parameters start to recover after ticular tissue, which is 5.7 times the EC50 reported
NANOTOXICOLOGY 11

by these authors. Moreover, at concentrations single intravenous injection of AgNPs (5 or 10 mg/


10 lg/ml, Ag-NPs induced a significant decline kg). Considering the higher dose used by
in SSC proliferation by disrupting components Gromadzka-Ostrowska et al. (2012), even if the
of the Glial cell-derived neurotrophic factor AgNPs were equally distributed through the various
(GDNF) signaling pathway, which is essential for body tissues/organs, which is probably not the
SSC self-renewal in vivo and in vitro (Braydich-Stolle case, the maximum amount of AgNPs reaching
et al. 2010). AgNPs was proven to be toxic for the each testicle would be 20 lg. Considering the local
cells involved in spermatogenesis, such as Leydig injection and the dose (100 lg/testicle) of AgNPs
and Sertoli cells and SSCs and this toxicity was used, we expected to observe more pronounced
linked to a rapid ROS production and accumulation changes in testicular morphology. Our findings sug-
(Zhang et al. 2015). High ROS levels in seminal gest that only spermied spermatozoa are affected,
plasma have been correlated with poor sperm either within the seminiferous tubules or in the epi-
morphology and is associated with the inhibition of didymis. Some reproductive toxicology studies clas-
sperm function and viability (Aziz et al. 2004). sified numerous chemicals that are toxic to the
However, in our study, we found the negative testis with the ability to alter the quantity/quality of
effects on male germinal cells are not long lasting, the sperm produced via spermatogenesis, together
suggesting that spermatogonia were not lethally with the quantity and quality of sperm that enter
impacted. In fact, most of the AgNP effects in the pre- the epididymis, thus identifying the epididymis as a
sent study seem to be on spermatid spermatozoa,
testis-independent target. Additionally, there are
which presented reduced motility and abnormal
numerous chemicals that produce epididymis-spe-
morphology. This may explain the normal parameters
cific reduction in cauda epididymal sperm number,
observed 56 days post-treatment, which correspond
without altering testicular sperm numbers (De
to a complete spermatogenesis process in rats
Grava Kempinas and Klinefelter 2014, 2018).
(Russell et al. 1990; McCarrey, 1993; Creasy, 1997).
The seminiferous tubule morphometric analysis
Gromadzka-Ostrowska et al. (2012) also suggested
showed that all the tubular parameters evaluated
that the genotoxicity effects of systemic AgNPs
significantly reduced in the AgNPs-D7 group,
observed in their study is pronounced in mature
whereas no difference was found between AgNPs-
sperm cells in the epididymis. Another in vitro study
D14, D28, and D56 and Control groups. Similar
with ejaculated sperm suggests AgNPs interfere with
results reported by Fathi et al. (2018), showed that
membrane receptors or cell signaling involved in
an intraperitoneal injection of AgNPs (125 mg/kg)
sperm motility (Terzuoli et al. 2012).
Regarding testicle histology, no damage or caused a significant reduction in the diameter, area
absence of any germ cell type was observed in the and circumference of seminiferous tubules. In con-
present study. Only a reduction in the amounts of trast, increased diameter, area and circumference of
spermatozoa and the presence of debris within the seminiferous tubules in animals treated with AgNPs
lumen of seminiferous tubules in the AgNPs-D7 were observed by Gromadzka-Ostrowska et al.
group were observed. Moreover, no spermatozoa (2012). Yet, Miresmaeili et al. (2013) found no sig-
were observed in the epididymis lumen of animals nificant changes in seminiferous tubule diameter in
in the AgNPs-D7 group, whereas some spermatozoa animals daily treated by the oral administration of
and debris were found in the epididymis of the AgNPs at doses varying from 25–200 mg/kg, after
AgNPs-D14 and AgNPs-D28 groups. Similarly, 48 days. These differences may be due to the differ-
Lafuente et al. (2016) reported an increased number ent administration routes, treatment period and
of epididymal sperm morphological abnormalities AgNP formulation employed. Moreover, in the pre-
but no significant changes in testicle and epididy- sent study, no differences between the relative
mis morphology of rats treated orally with PVP- weight of the testicles and epididymis in animals
AgNPs (50, 100, or 200 mg/kg) daily for 90 days. In treated with the AgNPs were observed. Similarly,
contrast, Gromadzka-Ostrowska et al. (2012) other studies using different AgNP administration
observed damage to the seminiferous epithelium, routes reported no significant relative changes in
wider intercellular spaces and vacuolization after a body, testicle or other organ weights in male rats
12 J. L. M. DE BRITO ET AL.

(Sung et al. 2009; Kim et al. 2010; Gromadzka- authors (Kim et al. 2010; Van Der Zande et al. 2012;
Ostrowska et al. 2012; Garcia et al. 2014). Yang et al. 2017) when administering AgNP to ani-
Regardless of the route of administration, NPs mals by other via (oral or intravenous), which might
can cross body barriers and enter the bloodstream be due to the very low dose we used (1.25 mg/
through the circulatory and lymphatic systems, kg) or a result of choosing an intratesticular admin-
translocating from the blood circulation to the main istration. In this work, minimal amounts of AgNPs
organs, such as liver, spleen, lungs, and kidneys were found in all the organs analyzed (liver, spleen,
(De Jong et al. 2008; Singh and Lillard 2009; lungs, and kidneys), but the percentage of silver
Dziendzikowska et al. 2012). Thus, we evaluated the found was consistently higher in the liver than in
animals for systemic functions and did not observe all of other organs examined. In fact, other authors
any alterations in water or food consumption. All described the liver as the target organ in AgNP tis-
animals gained weight as expected. Previous sue distribution (Kim et al. 2010; Lankveld et al.
reports showed that food and water consumption, 2010; Van Der Zande et al. 2012; Yang et al. 2017;
together with body and organ weight did not differ Gan et al. 2020), independent of the administration
between animals treated with AgNPs and control route. When administrated orally, silver is mostly
groups (Sung et al. 2009; Kim et al. 2010). excreted in feces, and the analysis of total silver
Moreover, all animals presented hematological showed accumulation in the liver (Jimenez-Lamana
parameters within the normal limits for Wistar rats et al. 2014). AgNPs are also excreted in feces when
(Cubas, Silva, and Cat~ao-Dias 2014). Some specific injected intravenously, and the authors suggested
parameters were altered, but without correlation to they were secreted by bile (Park et al. 2011).
the treatments. In the same way, other authors did Pathological changes in the liver together with sig-
not observe any significant changes in the hemato- nificant changes in serum AST and ALT were
logical parameters of rats exposed to AgNPs (Sung observed in mice receiving AgNP orally (250 mg/Kg)
et al. 2009; Kim et al. 2010; Yang et al. 2017). In this for 28 days, indicating that high doses of AgNPs
study, the biochemical parameters of ALT and cre- had a subacute toxicity on mice liver (Gan et al.
atinine were within the normal limits for Wistar rats 2020). Kim et al. (2010) also found damage to the
(Cubas, Silva, and Cat~ao-Dias 2014) for all animals, liver, including histopathological findings (bile-duct
while increased urea and AST values were observed hyperplasia and increased foci) and alkaline phos-
for all animals in the control, as well as in the phatase increase, when rats received 125 mg/kg of
treated groups, which are not correlated with the AgNP by gavage for 13 weeks. In the present study,
treatment. It is important to note that the literature ALS and AST levels did not suggest pathological
presents quite discrepant data for biochemical par- alterations in the liver of animals that received the
ameter limits in Wistar rats. For example, the AST intratesticular injection of AgNPs. It is important to
limits are described as 63–175 IU/l (Quesenberry note that the doses administered in the studies of
and Carpenter 2012) or 39–92 IU/l (Cubas, Silva, and Kim et al. (2010) and Gan et al. (2020) were much
Cat~ao-Dias 2014). Similarly, for urea, the limits are higher than the one used in the present study, and
described as 26–58 mg/dl (Lima et al. 2014) or that the animals received AgNP for several days. In
15–21 mg/dl (Cubas, Silva, and Cat~ao-Dias 2014). fact, no sign of any adverse effects was observed in
Other studies also related no significant alterations the animals in the present study.
in blood biochemical parameters in animals treated The intratesticular AgNP injection caused some
with AgNPs (Sung et al. 2009; Yang et al. 2017), acute and severe toxic effects in sperm cells,
although most of the reported studies did not use although not permanent, revealing reversibility
the same analytical approach. while not being able to neuter the treated animals.
We also used ICP-MS to analyze the presence of Despite that, no adverse effects are caused in the
silver in the liver, spleen, lungs, and kidneys after vital organs (liver, spleen, lungs, and kidneys) relat-
the intratesticular injection of AgNPs. In general, ing to the intratesticular injection of AgNPs. It is still
the amount of silver found in these organs (less possible that the intended animal neutering
than 200 ng/g of wet weight in any of the analyzed effect could be achieved by increasing the dose
organs) was lower than the described by other used. Using ICP-MS, it is possible to track the
NANOTOXICOLOGY 13

time-dependence (t) of the silver content (C) in key Ahmed, S. M., S. A. Abdelrahman, and S. M. Shalaby. 2017.
organs (liver, spleen, lungs, and kidneys) up to “Evaluating the Effect of Silver Nanoparticles on Testes of
Adult Albino Rats (Histological, Immunohistochemical and
56 days. Moreover, this study proposes a pioneering
Biochemical Study).” Journal of Molecular Histology 48 (1):
mathematical model describing C versus t, from 9–27. doi:10.1007/s10735-016-9701-4.
which the half-life time (Th) of the injected silver Amaku, M., R. A. Dias, and F. E. R. Ferreira. 2010. “Dynamics
(via AgNPs) in the liver (Th ¼ 17 days), spleen (Th ¼ and Control of Stray Dog Populations.” Mathematical
23 days), lungs (Th ¼ 30 days), and kidneys (Th ¼ Population Studies 17 (2): 69–78. doi:10.1080/
35 days) are successfully extracted. Considering the 08898481003689452
Ashoka, S., B. M. Peake, G. Bremner, K. J. Hageman, and
temporary effect of the AgNP intratesticular injec-
M. R. Reid. 2009. “Comparison of Digestion Methods for
tion and the absence of adverse effects, further ICP-MS Determination of Trace Elements in Fish Tissues.”
investigations on testicular injections of AgNPs Analytica Chimica Acta 653 (2): 191–199. doi:10.1016/j.aca.
must be done in the attempt to develop a contra- 2009.09.025.
ceptive method for male animals. Aziz, N., R. A. Saleh, R. K. Sharma, I. Lewis-Jones, N.
Esfandiari, A. J. Thomas, and A. Agarwal. 2004. “Novel
Association between Sperm Reactive Oxygen Species
Disclosure statement Production, Sperm Morphological Defects, and the Sperm
The authors report no conflict of interest. Deformity Index.” Fertility and Sterility 81 (2): 349–354. doi:
10.1016/j.fertnstert.2003.06.026.
Baki, M. E., S. M. Miresmaili, M. Pourentezari, E. Amraii, V.
Funding Yousefi, H. R. Spenani, A. R. Talebi, et al. 2014. “Effects of
Silver Nanoparticles on Sperm Parameters, Number of
This work was supported in part by the Coordenaç~ao de
Leydig Cells and Sex Hormones in Rats.” Iranian Journal of
Aperfeiçoamento de Pessoal de Nıvel Superior - Brasil
Reproductive Medicine 12 (2): 139–144.
(CAPES #1) under Grant (Finance Code 001); National
Braydich-Stolle, L. K., S. Hussain, J. J. Schlager, and M. C.
Institute of Science and Technology-Nanobiotechnology
Hofmann. 2005. “In Vitro Cytotoxicity of Nanoparticles in
(INCT-Nanobiotecnologia) of the Ministry of Science,
Mammalian Germline Stem Cells.” Toxicological Sciences :
Technology and Innovation (MCT/CNPq #2) under Grant
An Official Journal of the Society of Toxicology 88 (2):
(#573.880/2008-5); Ministry of Science and Innovation Spain
412–419. doi:10.1093/toxsci/kfi256.
(MICINN #3) under Grant (#MAT2017-88752-R); and J.L.M.
Braydich-Stolle, L. K., B. Lucas, A. Schrand, R. C. Murdock, T.
Brito received scholarships from CAPES and CNPq.
Lee, J. J. Schlager, S. M. Hussain, and M. Hofmann. 2010.
“Silver Nanoparticles Disrupt GDNF/Fyn Kinase Signaling
in Spermatogonial Stem Cells.” Toxicological Sciences : An
ORCID Official Journal of the Society of Toxicology 116 (2):
577–589. doi:10.1093/toxsci/kfq148.
Juliana Lis Mendes de Brito http://orcid.org/0000-0003- Caceres-Velez, P. R., M. L. Fascineli, M. H. Sousa, C. K. Grisolia,
3345-0449 L. Yate, P. E. N. Souza, I. Estrela-Lopis, S. Moya, and R. B.
Vanessa Nicolau de Lima http://orcid.org/0000-0003- Azevedo. 2018. “Humic Acid Attenuation of Silver
2836-1150 Nanoparticle Toxicity by Ion Complexation and the
Dorleta Otaegui Ansa http://orcid.org/0000-0002- Formation of a Ag3þ coating.” Journal of Hazardous
8215-354X Materials 353 (353): 173–181. doi:10.1016/j.jhazmat.2018.
Sergio Enrique Moya http://orcid.org/0000-0002- 04.019.
7174-1960 Creasy, D. M. 1997. “Evaluation of Testicular Toxicity in
Paulo Cesar Morais http://orcid.org/0000-0001-6181-7709 Safety Evaluation Studies: The Appropriate Use of
Ricardo Bentes de Azevedo http://orcid.org/0000-0002- Spermatogenic Staging.” Toxicologic Pathology 25 (2):
2137-9588 119–131. doi:10.1177/019262339702500201.
Carolina Madeira Lucci http://orcid.org/0000-0001- Cubas, Z. S., J. C. R. Silva, and J. L. Cat~ao-Dias. 2014. Tratado
7763-0746 de Animais Selvagens. S~ao Paulo: Editora Roca.
De Grava Kempinas, W., and G. R. Klinefelter. 2014.
“Interpreting Histopathology in the Epididymis.”
References
Spermatogenesis 4 (2): e979114. doi:10.4161/21565562.
ACC&D (Alliance for Contraception in Cats & Dog). 2016. 2014.979114.
ZeuterinTM/EsterilsolTM, Product profile and Position De Grava Kempinas, W., and G. R. Klinefelter. 2018. “The
Paper. http://www.acc-d.org/docs/default-source/Resource- Epididymis as a Target for Toxicants.” In Comprehensive
Library-Docs/pppp-zeuterinesterilsol-revisedmay-2016.pdf? Toxicology, edited by C.A. McQueen, 112–127. Oxford:
sfvrsn=2. p. 1–9. Elsevier.
14 J. L. M. DE BRITO ET AL.

De Jong, W. H., W. I. Hagens, P. Krystek, M. C. Burger, A. J. A. “Silver Nanoparticles Effects on Epididymal Sperm in Rats.”
M. Sips, and R. E. Geertsma. 2008. “Particle Size- Toxicology Letters 214 (3): 251–258. doi:10.1016/j.toxlet.
Dependent Organ Distribution of Gold Nanoparticles after 2012.08.028.
Intravenous Administration.” Biomaterials 29 (12): Jana, K., and P. K. Samanta. 2006. “Evaluation of Single
1912–1919. doi:10.1016/j.biomaterials.2007.12.037. Intratesticular Injection of Calcium Chloride for Nonsurgical
Dziendzikowska, K., J. Gromadzka-Ostrowska, A. Lankoff, M. Sterilization in Adult Albino Rats.” Contraception 73 (3):
Oczkowski, A. Krawczyn ska, J. Chwastowska, M. Sadowska- 289–300. doi:10.1016/j.contraception.2005.07.011.
Bratek, et al. 2012. “Time-Dependent Biodistribution and Jana, K., and P. K. Samanta. 2007. “Sterilization of Male Stray
Excretion of Silver Nanoparticles in Male Wistar Rats.” Dogs with a Single Intratesticular Injection of Calcium
Journal of Applied Toxicology: JAT 32 (11): 920–928. doi:10. Chloride: A Dose-Dependent Study.” Contraception 75 (5):
1002/jat.2758. 390–400. doi:10.1016/j.contraception.2007.01.022.
Ema, M., N. Kobayashi, M. Naya, S. Hanai, and J. Nakanishi. Jana, K., and P. K. Samanta. 2011. “Clinical Evaluation of
2010. “Reproductive and Developmental Toxicity Studies Non-Surgical Sterilization of Male Cats with Single Intra-
of Manufactured Nanomaterials.” Reproductive Toxicology Testicular Injection of Calcium Chloride.” BMC Veterinary
(Elmsford, N.Y.) 30 (3): 343–352. doi:10.1016/j.reprotox. Research 7 (1): 39. doi:10.1186/1746-6148-7-39.
2010.06.002. 
Jimenez-Lamana, J.,. F. Laborda, E. Bolea, I. Abad-Alvaro, J. R.
Fagundes, A. K.,F., E. C. S. Oliveira, B. M. Tenorio, C. C. S. Castillo, J. Bianga, H. M. Bierla, et al. 2014. “An Insight into
Melo, L. T. B. Nery, F. A. B. Santos, L. C. Alves, R. H. Silver Nanoparticles Bioavailability in Rats.” Metallomics 6
Douglas, and V. A. Silva. Jr, 2014. “Injection of a Chemical (12): 2242–2249. doi:10.1039/C4MT00200H.
Castration Agent, Zinc Gluconate, into the Testes of Cats Kar, A. B. 1961. “Chemical Sterilization of Male Rhesus mon-
Results in the Impairment of Spermatogenesis: A keys.” Endocrinology 69: 1116–1119. doi:10.1210/endo-69-
Potentially Irreversible Contraceptive Approach for This 6-1116.
Species?” Theriogenology 81 (2): 230–236. doi:10.1016/j. Kar, A. B., V. P. Kamboj, and A. Goswami. 1965. “Sterilization
theriogenology.2013.09.013. of Male Rhesus Monkeys by Iron Salts.” Journal of
FAO. 2011. Dog population management. Banna, Italy: FAO. Reproduction and Fertility 9 (1): 115–117. doi:10.1530/jrf.0.
http://www.fao.org/3/a-i4081e.pdf 0090115.
Fathi, N., S. M. Hoseinipanah, Z. Alizadeh, M. J. Assari, A. Kim, Y. S., M. Y. Song, J. D. Park, K. S. Song, H. R. Ryu, Y. H.
Moghimbeigi, M. Mortazavi, M. H. Hosseini, and M. Chung, H. K. Chang, et al. 2010. “Subchronic Oral Toxicity
Bahmanzadeh. 2018. “The Effect of Silver Nanoparticles on of Silver Nanoparticles.” Particle and Fibre Toxicology 7 (1):
the Reproductive System of Adult Male Rats: A 20. doi:10.1186/1743-8977-7-20.
Morphological, Histological and DNA Integrity Study.” Lafuente, D., T. Garcia, J. Blanco, D. J. Sanchez, J. J. Sirvent,
Advances in Clinical and Experimental Medicine 28 (3): J. L. Domingo, and M. Go mez. 2016. “Effects of Oral
299–305. doi:10.17219/acem/81607. Exposure to Silver Nanoparticles on the Sperm of Rats.”
Fordyce, G., P. B. Hodge, N. J. Beaman, A. R. Laing, C. Reproductive Toxicology (Elmsford, N.Y.) 60: 133–139. doi:
Campero, and Rk Shepherd. 1989. “An Evaluation of Calf 10.1016/j.reprotox.2016.02.007.
Castration by Intra-Testicular Injection of a Lactic Acid Lankveld, D. P. K., A. G. Oomen, P. Krystek, A. Neigh, A.
Solution.” Australian Veterinary Journal 66 (9): 272–276. Troost-de Jong, C. W. Noorlander, J. C. H. Van Eijkeren,
doi:10.1111/j.1751-0813.1989.tb13950.x. R. E. Geertsma, and W. H. De Jong. 2010. “The Kinetics of
Forzan, M. J., E. Garde, G. E. Perez, and R. V. Vanderstichel. the Tissue Distribution of Silver Nanoparticles of Different
2014. “Necrosuppurative Orchitis and Scrotal Necrotizing Sizes.” Biomaterials 31 (32): 8350–8361. doi:10.1016/j.bio-
Dermatitis following Intratesticular Administration of Zinc materials.2010.07.045.
Gluconate Neutralized with Arginine (EsterilSol) in 2 Leoci, R., G. Aiudi, V. Cicirelli, L. Brent, C. Iaria, and G. M.
Mixed-Breed Dogs.” Veterinary Pathology 51 (4): 820–823. Lacalandra. 2019. “Effects of Intratesticular vs
doi:10.1177/0300985813505875. Intraepididymal Calcium Chloride Sterilant on Testicular
Gan, J., J. Sun, X. Chang, W. Li, J. Li, S. Niu, L. Kong, et al. Morphology and Fertility in Dogs.” Theriogenology 127:
2020. “Biodistribution and Organ Oxidative Damage fol- 153–160. doi:10.1016/j.theriogenology.2019.01.006.
lowing 28 Days Oral Administration of Nanosilver with/ Levy, J. K., C. Crawford, L. D. Appel, and E. L. Clifford. 2008.
without Coating in Mice.” Journal of Applied Toxicology 40 “Comparison of Intratesticular Injection of Zinc Gluconate
(6): 815–817. doi:10.1002/jat.3946. versus Surgical Castration to Sterilize Male Dogs.”
Garcia, T. X., G. M. J. Costa, L. R. França, and M. C. Hofmann. American Journal of Veterinary Research 69 (1): 140–143.
2014. “Sub-Acute Intravenous Administration of Silver doi:10.2460/ajvr.69.1.140.
Nanoparticles in Male Mice Alters Leydig Cell Function Lima, C. M., A. K. Lima, M. G. D. Melo, G. A. A. Do ria, B. L. S.
and Testosterone Levels.” Reproductive Toxicology 45: Leite, M. R. Serafini, R. L. C. Albuquerque-J unior, and
59–70. doi:10.1016/j.reprotox.2014.01.006. A. A. S. Arau jo. 2014. “Valores de Refer^encia
Gromadzka-Ostrowska, J., K. Dziendzikowska, A. Lankoff, M. Hematolo gicos e Bioquımicos de Ratos (Rattus Novergicus
Dobrzyn ska, C. Instanes, G. Brunborg, A. Gajowik, J. Linhagem Wistar) Provenientes Do Bioterio da
Radzikowska, M. Wojewo dzka, and M. Kruszewski. 2012. Universidade Tiradentes.” Scientia Plena 10 (3): 1–9.
NANOTOXICOLOGY 15

Liu, Z., X. Liu, Y. Du, J. Ren, and X. Qu. 2015a. “Using Slater, M. R. 2001. “The Role of Veterinary Epidemiology in
Plasmonic Copper Sulfide Nanocrystals as Smart Light- the Study of Free-Roaming Dogs and Cats.” Preventive
Driven Sterilants.” ACS Nano 9(10): 10335–10346. doi:10. Veterinary Medicine 48 (4): 273–286. doi:10.1016/S0167-
1021/acsnano.5b04380. 5877(00)00201-4.
Liu, Z., X. Liu, X. Ran, E. Ju, J. Ren, and X. Qu. 2015b. “Single- Slater, M. R. 2002. Community Approaches to Feral Cats:
Layer Tungsten Oxide as Intelligent Photo-Responsive Problems, Alternatives, and Recommendations. Washington:
Nanoagents for Permanent Male Sterilization.” Biomaterials Humane Society of the US.
69: 56–64. doi:10.1016/j.biomaterials.2015.08.008. Slater, M. R., and S. Shain. 2005. “Feral Cats: An Overview.” In
Louvandini, H., C. McManus, R. D. Martins, C. M. Lucci, and The State of the Animals III, edited by D.J. Salem and A.N.
P. S. Corr^ea. 2008. “Caracterısticas Biometricas Testiculares Rowan, 43–53. Washington: Humane Society Press.
em Carneiros Santa In^es Submetidos a Diferentes Regimes Sung, J. H., J. H. Ji, J. D. Park, J. U. Yoon, D. S. Kim, K. S.
de Suplementaç~ao Proteica e Tratamentos anti- Jeon, M. Y. Song, et al. 2009. “Subchronic Inhalation
Helmınticos.” Ci^encia Animal Brasileira 9 (3): 638–647. Toxicity of Silver Nanoparticles.” Toxicological Sciences: An
McCarrey, J. 1993. “Development of the Germ Cell.” In Cell Official Journal of the Society of Toxicology 108 (2):
and Molecular Biology of the Testis, edited by C. Desjardins 452–461. doi:10.1093/toxsci/kfn246.
and L. Ewing, 58–89. New York: Oxford University Press. Terzuoli, G., F. Iacoponi, E. Moretti, T. Renieri, G. Baldi, and G.
Miresmaeili, S. M., I. Halvaei, F. Fesahat, A. Fallah, N. Collodel. 2012. “Vitro Effect of Silver Engineered
Nikonahad, and M. Taherinejad. 2013. “Evaluating the Role Nanoparticles on Human Spermatozoa.” Journal of the
of Silver Nanoparticles on Acrosomal Reaction and Siena Academy of Sciences 3 (1): 27–29. doi:10.4081/jsas.
Spermatogenic Cells in Rat.” Iranian Journal of 2011.27.
Reproductive Medicine 11 (5): 423–430. Van Der Zande, M., R. J. Vandebriel, E. Van Doren, E. Kramer,
Oliveira, E. C. S., A. K. F. Fagundes, C. C. S. Melo, L. T. B. Nery, Z. Herrera Rivera, C. S. Serrano-Rojero, E. R. Gremmer,
R. G. R^evoredo, T. G. F. Andrade, K. Oliveira-Esquerre, J. P.
et al. 2012. “Distribution, Elimination, and Toxicity of Silver
Kastelic, and V. A. Silva. Jr, 2013. “Intratesticular Injection
Nanoparticles and Silver Ions in Rats after 28-Day Oral
of a Zinc-Based Solution for Contraception of Domestic
Exposure.” ACS Nano 6 (8): 7427–7442. doi:10.1021/
Cats: A Randomized Clinical Trial of Efficacy and Safety.”
nn302649p.
The Veterinary Journal 197 (2): 307–310. doi:10.1016/j.tvjl.
Vannucchi, Ci, D. S. R. Angrimani, A. R. Eyherabide, C. P.
2013.01.011.
Mazzei, C. F. Lucio, P. C. Maiorka, L. C. G. Silva, and M.
Olson, P. N., and S. D. Johnston. 1993. “New Developments
Nichi. 2015. “Effects of Intratesticular Administration of
in Small Animal Population Control.” Journal of American
Zinc Gluconate and Dimethyl Sulfoxide on Clinical,
Veterinary Association 202 (6): 904–909.
Endocrinological, and Reproductive Parameters in Dogs.”
Park, K., E. J. Park, I. K. Chun, K. Choi, S. H. Lee, J. Yoon, and
Theriogenology 84 (7): 1103–1110. doi:10.1016/j.therioge-
B. C. Lee. 2011. “Bioavailability and Toxicokinetics of
nology.2015.06.005.
Citrate-Coated Silver Nanoparticles in Rats.” Archives of
Wiebe, J. P., K. J. Barr, and K. D. Buckingham. 1989.
Pharmacal Research 34 (1): 153–158. doi:10.1007/s12272-
“Sustained Azoospermia in Squirrel Monkey, Saimiri
011-0118-z.
Quesenberry, K. E., and J. W. Carpenter. 2012. Ferrets, Sciureus, Resulting from a Single Intratesticular Glycerol
Rabbits, and Rodents Clinical Medicine and Surgery. St Injection.” Contraception 39 (4): 447–457. doi:10.1016/
Louis: Elsevier. 0010-7824(89)90122-4.
Rafatmah, D., A. Mogheiseh, and D. Eshghi. 2019. “Chemical Yang, L., H. Kuang, W. Zhang, Z. P. Aguilar, H. Wei, and H.
Sterilization with Intratesticular Administration of Zinc Xu. 2017. “Comparisons of the Biodistribution and
Gluconate in Adult Dogs: A Preliminary Report.” Basic and Toxicological Examinations after Repeated Intravenous
Clinical Andrology 29: 12. doi:10.1186/s12610-019-0092-8. Administration of Silver and Gold Nanoparticles in Mice.”
Russell, L. D., R. A. Ettlin, A. P. S. Hikim, and E. D. Clegg. Scientific Reports 7 (1): 3303. doi:10.1038/s41598-017-
1990. Histological and Histopathological Evaluation of the 03015-1.
Testis. Florida: Cache River Press. Yostawonkul, J., S. Surassmo, K. Namdee, M. Khongkow, C.
Silva, R. C. A., C. S. Paranzini, L. G. Franco, M. P. Miguel, C. S. Boonthum, S. Pagseesing, N. Saengkrit, et al. 2017. “
Honsho, and F. F. Souza. 2018. “Calcium Chloride Nanocarrier-Mediated Delivery of a-Mangostin for Non-
Combined with Dimethyl Sulphoxide for the Chemical Surgical Castration of Male Animals.” Scientific Reports 7
Sterilization of dogs.” Reproduction in Domestic (1): 16234. doi:10.1038/s41598-017-16563-3.
Animals ¼ Zuchthygiene 53 (6): 1330–1338. doi:10.1111/rda. Zhang, X. F., Y. J. Choi, J. W. Han, E. Kim, J. H. Park, S.
13252. Gurunathan, and J. H. Kim. 2015. “Differential
Singh, R., and J. W. Lillard. 2009. “Nanoparticle-Based Nanoreprotoxicity of Silver Nanoparticles in Male Somatic
Targeted Drug Delivery.” Experimental and Molecular Cells and Spermatogonial Stem cells.” International Journal
Pathology 86 (3): 215–223. doi:10.1016/j.yexmp.2008.12. of Nanomedicine 10 (1): 1335–1357. doi:10.2147/IJN.
004. S76062.

You might also like