Invest Clin 66(4): 408 - 425, 2025 https://doi.org/10.54817/IC.v66n4a05
Corresponding author: Dandan Qiu. Department of Urology, The First Affiliated Hospital of Zhejiang Chinese
Medical University, Zhejiang Hospital of Traditional Chinese Medicine, Hangzhou Zhejiang, 310006, China. Phone:
+8613588454495. Email: qiudandan2000@outlook.com
Rutin as a potential therapeutic agent
for arsenic-induced testicular damage:
Antioxidant, anti-inflammatory,
and anti-apoptotic effects.
Yunyun Wang1,#, Zengrong Hua2,#, Jin Qing Hui3 and Dandan Qiu4
1Department of Pharmacy, The First Aliated Hospital of Zhejiang Chinese Medical
University, Zhejiang Hospital of Traditional Chinese Medicine, Hangzhou, Zhejiang, China.
2Department of Urology, Huai'an Clinical Medical College of Jiangsu University
(Huai'an Hospital of Huai'an City), Huai'an, Jiangsu, China.
3Department of Surgical, Shaanxi Kangfu Hospital, Xian, Shaanxi, China.
4Department of Urology, The First Affiliated Hospital of Zhejiang Chinese Medical
University, Zhejiang Hospital of Traditional Chinese Medicine, Hangzhou Zhejiang, China.
#Yunyun Wang and Zengrong Hua are co-first authors; they contributed equally
to this work.
Keywords: Apoptosis; Arsenic; Inflammation; Rutin, Spermatogenesis; Testicular
Toxicity.
Abstract. Arsenic exposure is associated with male reproductive toxicity, in-
cluding impaired spermatogenesis, reduced sperm quality, and disrupted hormone
levels. Rutin, a natural flavonoid, has demonstrated protective effects in various or-
gans owing to its antioxidant, anti-inflammatory, and antiapoptotic properties. This
study aimed to assess the efficacy of rutin in ameliorating testicular toxicity caused
by sodium arsenite in laboratory rats. Sprague-Dawley rats were randomly assigned
to normal, arsenic control, rutin (25, 50, and 100 mg/kg), and Co-enzyme Q10 (10
mg/kg) treatment groups. Testicular damage was induced by oral administration of
sodium arsenite (10 mg/kg, for two days). Rutin and Co-enzyme Q10 were adminis-
tered orally for 15 and seven days, respectively. The serum hormone levels, sperm pa-
rameters, testicular mitochondrial enzymes, oxidative stress markers, inflammatory
cytokines, apoptotic proteins, and histopathology were assessed. Arsenic exposure
significantly decreased (p<0.001) sperm parameters (count, motility, and viabil-
ity), serum hormone levels (follicle-stimulating hormone, luteinizing hormone, and
testosterone), and mitochondrial enzyme activity. Rutin (50 and 100 mg/kg) sig-
nificantly (p<0.01 and p<0.001) attenuated arsenic-induced alterations in a dose-
dependent manner, improving organ weight, sperm parameters, and hormone lev-
els. Rutin also improved mitochondrial complex activity and testicular architecture.
In contrast, elevated oxidative stress (reduced glutathione, superoxide dismutase,
nitric oxide, and lipid peroxidation), inflammation (tumor necrosis factor-alpha,
Protective effects of rutin against arsenic-induced testicular damage 409
Vol. 66(4): 408 - 425, 2025
Rutina, un agente terapéutico potencial para el daño testicular
inducido por arsénico: Efecto antioxidante, antinflamatorio,
y anti-apoptótico.
Invest Clin 2025; 66 (4): 408 – 425
Palabras clave: Apoptosis; Arsénico; Inflamación; Rutina; Espermatogénesis; Toxicidad
Testicular.
Resumen. La exposición al arsénico se asocia con toxicidad reproductiva mas-
culina que incluye alteración de la espermatogénesis, reducción de la calidad del
esperma y alteración de los niveles hormonales. La rutina, un flavonoide natural, ha
demostrado efectos protectores en varios órganos debido a sus propiedades antio-
xidantes, antiinflamatorias y antiapoptóticas. El propósito del trabajo fue evaluar la
eficacia de la rutina para reducir la toxicidad testicular causada por el arsenito de
sodio en ratas de laboratorio. Ratas (Sprague-Dawley) fueron asignadas aleatoria-
mente a grupos de tratamiento normal, control de arsénico, rutina (25, 50 y 100
mg/kg) y coenzima Q10 (10 mg / kg). El daño testicular se indujo mediante la ad-
ministración oral de arsenito de sodio (10 mg/kg, 2 días). La rutina y la coenzima
Q10 se administraron por vía oral durante 15 y 7 días, respectivamente. Se evalua-
ron los niveles séricos de hormonas, los parámetros de los espermatozoides, las enzi-
mas mitocondriales testiculares, los marcadores de estrés oxidativo, las citocinas in-
flamatorias, las proteínas apoptóticas y la histopatología. La exposición al arsénico
disminuyó significativamente (p<0,001) los parámetros espermáticos (recuento,
motilidad y viabilidad), y los niveles séricos de hormonas (hormona estimulante del
folículo, hormona luteinizante y testosterona) y la actividad de las enzimas mitocon-
driales. La rutina (50 y 100 mg/kg) atenuó significativamente (p<0,01 y p<0,001)
las alteraciones inducidas por el arsénico de manera dosis-dependiente, mejorando
el peso de los órganos, los parámetros espermáticos y los niveles hormonales. La
rutina también mejoró la actividad del complejo mitocondrial y la arquitectura tes-
ticular, mientras que el estrés oxidativo elevado (glutatión reducido y superóxido
dismutasa, óxido nítrico y peroxidación lipídica), la inflamación (factor de necrosis
tumoral alfa, interleucina-6 e interleucina-1β) y la apoptosis (expresión de proteínas
caspasa-3 y caspasa-9) mejoraron con la rutina. En conclusión, la rutina demostró
efectos protectores significativos contra la toxicidad testicular inducida por el ar-
senito de sodio en ratas al reducir el estrés oxidativo, la inflamación y la apoptosis.
Estos hallazgos sugieren que la rutina tiene potencial terapéutico para mitigar la
toxicidad reproductiva asociada al arsénico.
Received: 12-08-2025 Accepted: 22-09-2025
interleukin-6, and interleukin-1β), and apoptosis (caspase-3 and caspase-9 protein
expression) were ameliorated by rutin. In conclusion, rutin demonstrated signifi-
cant protective effects against sodium arsenite-induced testicular toxicity in rats by
reducing oxidative stress, inflammation, and apoptosis. These findings suggest that
rutin has therapeutic potential in mitigating arsenic-related reproductive toxicity.
410 Wang et al.
Investigación Clínica 66(4): 2025
INTRODUCTION
Arsenic is a metalloid commonly found
in the environment, known for its toxicity
and important public health effects 1,2. It
mainly impacts populations through con-
taminated drinking water, often from natural
sources and human activities such as indus-
trial discharge and the use of arsenic-based
pesticides 3,4. According to the World Health
Organization (WHO), more than 140 million
people worldwide are exposed to arsenic-
contaminated water exceeding the safe limit
of 10 µg/L 5. Furthermore, long-term expo-
sure to arsenic has been associated with nu-
merous health problems, including cancer
and various non-cancerous conditions like
cardiovascular diseases, diabetes, and repro-
ductive issues 6,7. Sodium arsenite is recog-
nized for causing testicular toxicity through
mechanisms such as oxidative stress, apop-
tosis, and inflammation. Researchers believe
that inflammation, oxidative/nitrosative
stress, and apoptosis are key factors in arse-
nic-induced testicular damage 6,8.
The impact of arsenic toxicity on re-
productive health has become an increas-
ing concern, particularly regarding the male
reproductive system 9. Arsenic exposure is
linked to various reproductive problems,
including male infertility, decreased sperm
quality, and disruptions in testosterone pro-
duction. This results in reduced weights of
reproductive organs, more sperm abnormali-
ties, and apoptosis in testicular cells 10. The
harmful effects of arsenic on male fertility
involve several mechanisms, such as inter-
ference with steroidogenesis and mitochon-
drial dysfunction in reproductive tissues 9.
These disruptions lead to lower testoster-
one levels, decreased sperm count, and ab-
normal sperm morphology 11. Additionally,
changes in spermatogenesis and reduced
gonadotropin levels have been observed, fur-
ther emphasizing the reproductive risks of
arsenic 3. Moreover, arsenic exposure causes
increased production of reactive oxygen spe-
cies (ROS), indicating the role of oxidative
stress in its genotoxic effects 12. Research-
ers have documented that testicular toxicity
caused by sodium arsenite is partly due to its
ability to induce apoptosis through the p53
pathway, as shown in zebrafish studies where
sodium arsenite exposure elevated apoptosis
markers and decreased global DNA methyla-
tion 13. Furthermore, experiments with Cae-
norhabditis elegans demonstrated that sodi-
um arsenite can cause cell cycle arrest and
germline apoptosis, both of which depend on
concentration and exposure time 12.
Several protective agents have been stud-
ied to reduce testicular toxicity caused by so-
dium arsenite. Melatonin has been shown to
decrease arsenic-induced testicular cell death,
oxidative stress, and tissue damage by boost-
ing antioxidant enzyme activity and lowering
lipid peroxidation 14. Similarly, Salvia hispani-
ca (chia seeds) proved effective in decreasing
testicular toxicity by improving sperm quality,
serum sex hormone levels, and antioxidant en-
zyme activity, thanks to its flavonoid content
and antioxidant properties 15. Hesperidin and
lipoic acid were also tested for their protec-
tive effects against liver, kidney, and testicular
toxicity when administered with sodium arse-
nite16. Rutin is a natural flavonoid with a wide
range of pharmacological benefits, including
antioxidant, antimicrobial, antifungal, anti-
allergic, anti-cancer, anti-inflammatory, and
antiapoptotic effects 4,17-22. Additionally, rutin
has been shown to help manage neurodegen-
erative diseases and metabolic conditions like
diabetes due to its cell-protective effects 19. Its
protective properties have been documented
in the liver, kidneys, and heart due to its anti-
inflammatory, antioxidant, and antiapoptotic
actions, making it a candidate for protecting
these organs from toxic agents 4. However, the
impact of rutin on testicular toxicity caused
by sodium arsenite has not yet been studied.
Given its protective effects on other organs, ru-
tin might provide similar benefits in reducing
testicular damage from sodium arsenite. This
study examined the effectiveness of rutin in al-
leviating testicular toxicity caused by sodium
arsenite in male rats.
Protective effects of rutin against arsenic-induced testicular damage 411
Vol. 66(4): 408 - 425, 2025
MATERIALS AND METHODS
Experimental design
Rats (male, Sprague-Dawley, weighing
200-220 g, sourced from the animal house
of Shaanxi Kangfu Hospital, China) were
randomly divided into the following groups
(n=6 per group): normal, arsenic control
(As), rutin (Sigma Chemical Co., St Louis,
Missouri, United States; 25, 50, and 100
mg/kg), and Co-enzyme Q10 (Medicines Pvt.
Ltd. Mumbai, India; 10 mg/kg). Testicular
damage was induced in rats (except the nor-
mal group) by oral administration of sodium
arsenite (Otto Chemicals, India; 10 mg/kg,
for two days) 23,24. Rats in the arsenic control
and normal groups were treated with double-
distilled water (10 mL/kg). Three different
dosages of rutin (25, 50, and 100 mg/kg)
were selected based on previous studies 21,22
and were administered orally for 15 days. Co-
enzyme Q10 was administered for seven con-
secutive days before arsenite administration
and continued for up to 15 days. On the 16th
day, the rats were put under anesthesia us-
ing ether, and blood was drawn through ret-
ro-orbital puncture. Each blood sample was
placed in a separate vial for analysis of se-
rum parameters. Serum follicle-stimulating
hormone, luteinizing hormone, and testos-
terone levels were quantified according to
the manufacturer’s instructions for the rat-
specific ELISA kit (Thermo Scientific, Rock-
ford, IL, USA). The rats were euthanized by
carbon dioxide asphyxiation, and the testes
were rapidly removed and stored at -80°C for
biochemical parameters. Other organs, such
as the epididymis and prostate, were isolated
and weighed. The epididymal sperm count
and motility were determined according to a
previously reported method 25.
Biochemical estimation of testis
homogenate
Supernatant of the tissue homogenate
was employed to estimate lipid peroxidation
[malondialdehyde (MDA) content], nitric
oxide (NO content), reduced glutathione
(GSH), and superoxide dismutase (SOD)]
as described previously 26-29. Testicular mito-
chondrial enzyme activities, including Com-
plex I (nicotinamide-adenine dinucleotide
(NADH) dehydrogenase activity), Complex
II (succinate dehydrogenase (SDH) activity),
Complex III (mitochondrial redox activity),
and Complex IV (cytochrome oxidase assay)
were estimated according to previously re-
ported methods 30,31.
Testicular interleukins (interleukins
(ILs); IL-6 (ERA31RB) and IL-1β (BMS630))
and tumor necrosis factor-alpha (TNF-α;
ERA56RB) were quantified in the testis ho-
mogenate using Enzyme-linked immunosor-
bent assay (ELISA) kits (Thermo Scientific,
Rockford, IL, USA). Briefly, 500 mg of testis
tissue samples were homogenized with a me-
chanical homogenizer in 5 ml of phosphate-
buffered saline at 3000 rpm. The homoge-
nate was centrifuged for 30 min at 20,000
rpm (4°C) in a cryo centrifuge (Eppendorf),
and the supernatant was used to determine
ILs and TNF-α. Briefly, the quantification of
ILs and TNF-α was performed using the Ther-
mo Scientific Rat ILs and TNF-α immunoas-
say kit, following the instructions provided.
The Rat ILs and TNF-α immunoassay was a
4.5 h solid phase designed to measure rat
ILs and TNF-α levels. The assay employed a
sandwich enzyme immunoassay principle. A
monoclonal antibody specific for rat ILs and
TNF-α was pre-coated on the microplates.
Standards, control, and samples were pipet-
ted into the wells, and the immobilized an-
tibody thus bound any rat ILs or TNF-α pres-
ent in the sample. After washing away the
unbound substance, an enzyme-linked poly-
clonal antibody specific for rat ILs or TNF-α
was pipetted into the microtitre wells. Any
unbound antibody was washed off, and then
a substrate solution was added to the wells.
The enzymatic reaction produced a blue
product that turned yellow upon addition of
the stop solution. The intensity of the gener-
ated color was measured and was proportion-
al to the amount of rat ILs or TNF-α bound
in the initial steps. A standard curve was run
412 Wang et al.
Investigación Clínica 66(4): 2025
on each assay plate using recombinant ILs or
TNF-α in serial dilutions. The sample values
were then read and calculations made ac-
cording to the standard curve. Values were
expressed as means ± S.E.M. The levels of
ILs or TNF-α were expressed as units per mg
of gastric tissue.
The protein expression of caspase-3 and
caspase-9 was assessed by western blot in the
testis 32-34. Briefly, the testis was sonicated in
Tissue Protein Extraction Reagent (Thermo
Fisher Scientific, Inc., Mumbai, Maharash-
tra, India). The lysates were centrifuged at
10,000 × g for 10 min at 4°C. Protein con-
centration was determined using a Bicin-
choninic Acid (BCA) assay kit (Beyotime
Shanghai, China) on ice for 30 min. Equal
amounts of extracted protein samples (50
µg) were separated by 10% SDS-PAGE (sodi-
um dodecyl sulfate-polyacrylamide gel elec-
trophoresis) and transferred onto polyvinyli-
dene difluoride membranes. The membranes
were blocked with 5% non-fat dry milk at
37°C for 1 hr and incubated overnight at 4°C
with the primary antibodies recognizing cas-
pase-3 (ab4051; 1/200 dilution; 31 kDa) and
caspase-9 (ab202068; 1/2000 dilution; 46
kDa; Abcam, Cambridge, MA, USA). In ad-
dition, an anti-rabbit horseradish-linked sec-
ondary antibody (goat anti-rabbit IgG H&L;
ab97051) was used, which was incubated at
37°C for 2 hr. Protein bands were visualized
using the Chemiluminescent kit (Bio-Rad
Laboratories, Inc., Mumbai, Maharashtra,
India), and glyceraldehyde 3-phosphate de-
hydrogenase (GAPDH; EPR6256, ab128915;
1/10000 dilution; 36 kDa) served as the
loading control.
Histopathological analysis of testis
Other testis samples were preserved in
10% formalin for 24 hours. These samples
underwent dehydration and were immersed
in xylene for one hour, repeated three times,
followed by treatment with ethyl alcohol at
concentrations of 70%, 90%, and 100% for
two hours. The infiltration and impregnation
process involved treating the samples with
paraffin wax twice, with each session lasting
1 hour. The tissue samples were then sliced
into sections with a thickness of 3-5µm and
stained using hematoxylin and eosin (H&E).
The sections were mounted on slides with
Distrene Pthalate Xylene (DPX) serving as
the mounting medium. A light microscope
was used to examine the sections for histo-
pathological characteristics and cell infiltra-
tion. The observed changes in histological
features were categorized into grades rang-
ing from 0 to 4 according to a previously re-
ported method 35.
Statistical analysis
The data are presented as mean ±
standard error of the mean (SEM). Graph-
Pad Prism 5.0 software (GraphPad, San Di-
ego, CA, USA) was utilized for data analysis.
Biochemical parameter data were examined
using one-way analysis of variance, followed
by Tukey’s multiple range test for paramet-
ric results, while the Kruskal-Wallis test was
used for non-parametric outcomes. A p of
less than 0.05 was deemed statistically sig-
nificant. Correlation coefficients were calcu-
lated using a two-sided Fisher’s test.
RESULTS
Attenuation of arsenic-induced alteration
in body weight and organ weights by rutin
Table 1 presents the descriptive results
for various body and organ weight parame-
ters across the different treatment groups.
The arsenic (As) control group had a signifi-
cant reduction (p<0.001) in organ weights
(testes, epididymis, and prostate) and body
weight compared with the normal group.
CoQ treatment resulted in a significant im-
provement (p<0.001) in body weight and
organ weights compared to the arsenic con-
trol group. Rutin (50 and 100 mg/kg) ad-
ministration also effectively (p<0.01 and
p<0.001) and dose-dependently increased
the body, testes, epididymis, and prostate
weights relative to the arsenic-exposed
group.
Protective effects of rutin against arsenic-induced testicular damage 413
Vol. 66(4): 408 - 425, 2025
Table 1. Attenuation of arsenic-induced alteration in body weight, organ weights (testes, epididymis, and prostate),
and sperm parameters (count, motility, and viability) by rutin.
Parameters Normal As Control CoQ (10) R (25) R (50) R (100)
Body weight (gm) 281.8 ± 2.86 217.50 ± 3.73### 275.70 ± 2.69*** 214.70 ± 2.80 247.00 ± 4.02** 262.20 ± 2.77***
Testes weight (gm) 1.79 ± 0.04 1.03 ± 0.07### 1.59 ± 0.04*** 1.07 ± 0.05 1.51 ± 0.06** 1.68 ± 0.02***
Testes weight/Body weight (×10-3) 6.34 ± 0.15 4.75 ± 0.39### 5.76 ± 0.18*** 4.99 ± 0.21 6.13 ± 0.33** 6.41 ± 0.12***
Epididymis weight (gm) 0.51 ± 0.01 0.19 ± 0.02### 0.45 ± 0.02*** 0.23 ± 0.01 0.33 ± 0.01** 0.45 ± 0.01***
Epididymis weight/Body weight (×10-3) 1.80 ± 0.03 0.87 ± 0.07### 1.62 ± 0.07*** 1.09 ± 0.07 1.32 ± 0.04** 1.73 ± 0.04***
Prostate weight (mg) 612.30 ± 10.94 308.30 ± 9.78### 560.70 ± 9.02*** 298.70 ± 11.44 478.30 ± 11.78** 541.70 ± 11.45***
Prostate weight/Body weight (×10-3) 2.18 ± 0.04 1.42 ± 0.05### 2.03 ± 0.04*** 1.39 ± 0.04 1.94 ± 0.07** 2.07 ± 0.05***
Sperm count (millions/mL) 60.17 ± 1.25 34.00 ± 0.97### 56.83 ± 1.30*** 33.83 ± 0.75 45.00 ± 1.21** 51.83 ± 1.25***
Sperm motility (%) 71.33 ± 1.05 40.33 ± 1.02### 62.00 ± 1.00*** 44.33 ± 0.95 49.17 ± 1.49** 59.50 ± 0.67***
Dead sperm (%) 28.67 ± 1.05 59.67 ± 1.02### 38.00 ± 1.00*** 55.67 ± 0.95 50.83 ± 1.49** 40.50 ± 0.67***
Abnormal sperm (%) 12.17 ± 0.60 36.50 ± 0.43### 17.17 ± 0.54*** 34.17 ± 0.48 24.67 ± 0.42** 19.33 ± 0.76***
Data analysis: One-way ANOVA (post-hoc test: Tukey’s multiple range test). Data are reported as mean ± SEM (n=6). Statistically significant compared with
###normal rats, ** and ***as control rats. ###p < 0.001, **p < 0.01 and ***p < 0.001. Arsenic (As), Co-enzyme Q10 (CoQ (10)) and Rutin (R).
Table 2. Attenuation of arsenic-induced alteration in serum luteinizing hormone, follicle stimulating hormone,
testosterone levels and testicular mitochondrial complex activities by rutin.
Parameters Normal As Control CoQ (10) R (25) R (50) R (100)
Serum parameters
Luteinizing hormone (ng/mL) 1.88 ± 0.04 0.26 ± 0.05### 0.33 ± 0.03 0.36 ± 0.04 0.71 ± 0.05** 1.21 ± 0.04***
Follicle stimulating hormone (ng/mL) 1.37 ± 0.02 0.16 ± 0.02### 0.15 ± 0.02 0.26 ± 0.02 0.64 ± 0.02** 0.92 ± 0.02***
Testosterone (ng/mL) 3.69 ± 0.04 1.41 ± 0.05### 1.32 ± 0.06 1.5 ± 0.06 2.39 ± 0.04** 2.86 ± 0.05***
Testicular parameters
Complex I (nmole of NADH oxidized/
min/mg protein) 33.36 ± 1.66 7.41 ± 1.80### 30.21 ± 1.79*** 10.07 ± 1.35 16.91 ± 1.25** 28.31 ± 2.10***
Complex II (mmole/mg protein) 14.35 ± 0.82 4.38 ± 0.78### 11.86 ± 0.84*** 4.74 ± 0.62 9.42 ± 0.67** 11.24 ± 0.58***
MTT assay (OD at 540 nm) 0.45 ± 0.03 0.17 ± 0.03### 0.45 ± 0.03*** 0.21 ± 0.02 0.32 ± 0.01** 0.39 ± 0.02***
Complex-IV (nmol cyto-C oxidized/
min/mg protein) 6319.00 ± 280.30 1008.00 ± 212.70### 5293.00 ± 254.70*** 1586.00 ± 191.90 3224.00 ± 201.30** 4515.00 ± 342.00***
Data analysis: One-way ANOVA (post-hoc test: Tukey’s multiple range test). Data are reported as mean ± SEM (n=6). Statistically significant compared with
###normal rats, ** and ***as control rats. ###p < 0.001, **p < 0.01 and ***p < 0.001. Arsenic (As), Co-enzyme Q10 (CoQ (10)), 3-(4,5-dimethylthiazol-2-yl)-2,5-
diphenyltetrazolium bromide (MTT) and Rutin (R).
414 Wang et al.
Investigación Clínica 66(4): 2025
Attenuation of arsenic-induced alteration
in sperm parameters by rutin
The arsenic control group showed a
significant reduction (p<0.001) in cell
count and motility compared to the con-
trol group. However, the percentage of dead
and abnormal sperm was markedly higher
(p<0.001) in the arsenic control group
than that in the normal group. Treatment
with CoQ resulted in significant improve-
ments (p<0.001) in sperm count and mo-
tility compared with the arsenic control
group. Furthermore, CoQ administration
resulted in a more effective (p<0.001) re-
covery of the percentage of dead and abnor-
mal sperm than arsenic control. Rutin (50
and 100 mg/kg) treatment demonstrated a
marked (p<0.01 and p<0.001) and dose-
dependent improvement in sperm param-
eters relative to the arsenic-exposed group
(Table 1).
Attenuation of arsenic-induced alteration
in serum follicle-stimulating hormone,
luteinizing hormone, and testosterone
levels by rutin
Exposure to sodium arsenite significant-
ly decreased (p<0.001) the levels of serum
follicle-stimulating hormone, luteinizing hor-
mone, and testosterone in the arsenic con-
trol group compared with the normal group.
Treatment with CoQ and rutin (25 mg/kg)
had a minimal impact on serum luteinizing
hormone, follicle-stimulating hormone, and
testosterone hormone levels. However, rutin
(50 and 100 mg/kg) treatments demonstrat-
ed effective, dose-dependent increases in all
three hormones (p<0.01 and p<0.001, re-
spectively) compared with the arsenic control
group (Table 2).
Attenuation of arsenic-induced alteration
in testicular mitochondrial complex
activities by rutin
Arsenic exposure led to a significant
reduction (p<0.001) in mitochondrial com-
plex activity compared to the normal group.
The results presented in Table 2 demonstrate
that both CoQ and rutin (50 and 100 mg/
kg) significantly improved mitochondrial
function in arsenic-exposed rats. Specifical-
ly, rutin (50 and 100 mg/kg) administration
led to notable (p<0.01 and p<0.001) and
dose-dependent increases in mitochondrial
complex activities compared with the arse-
nic control group. However, rutin (25 mg/
kg) did not show any significant potential to
improve mitochondrial function or mitigate
arsenic-induced damage.
Attenuation of arsenic-induced alteration
in testicular oxido-nitrosative stress
by rutin
The results presented in Table 3 show
the effects of rutin treatment on testicular
oxido-nitrosative stress. Arsenic exposure
caused a substantial (p<0.001) decrease in
GSH and SOD levels and a marked (p<0.001)
elevation in nitric oxide and MDA levels in
the arsenic control group compared to the
normal group. CoQ treatment significantly
(p<0.001) improved GSH and SOD levels
and effectively reduced (p<0.001) nitric ox-
ide and MDA levels compared with the arse-
nic control group. Rutin (25 mg/kg) treat-
ment showed minimal effects in attenuating
arsenic-induced elevated testicular oxido-
nitrosative stress compared with the arsenic
control group. However, rutin (50 and 100
mg/kg) demonstrated marked (p<0.01 and
p<0.001) and dose-dependent protective ef-
fects, reflected by restored antioxidant sta-
tus (GSH and SOD levels) and reduced oxi-
dative stress markers (nitric oxide and MDA)
to normal levels (Table 3).
Attenuation of arsenic-induced alteration
in testicular inflammatory markers
activities by rutin
The results in Table 3 show significant
differences in inflammatory cytokine lev-
els between the normal and arsenic control
groups. The arsenic control group exhibited
markedly elevated (p<0.001) levels of ILs
(IL-6 and IL-1β) and TNF compared with
Protective effects of rutin against arsenic-induced testicular damage 415
Vol. 66(4): 408 - 425, 2025
the normal group. CoQ treatment substan-
tially (p<0.001) lowered ILs (IL-6 and IL-
1β) and TNF levels compared to the arse-
nic control group. Rutin (50 and 100 mg/
kg) administration resulted in effective
(p<0.01 and p<0.001) and dose-dependent
reductions in cytokine levels compared with
the arsenic control group. However, a lower
dose of rutin (25 mg/kg) did not reduce cy-
tokine levels to levels comparable to those of
the arsenic control group, indicating failure
to ameliorate arsenic-induced inflammation
(Table 3).
Attenuation of arsenic-induced alteration
in testicular caspase-3 and caspase-9
protein expressions by rutin
Fig. 1 illustrates the impact of arsenic
exposure on markers of apoptosis in the tes-
tes, along with their correlation with sperm
count. The caspase-3 and caspase-9 pro-
tein expression was significantly increased
(p<0.001) in the arsenic control group
compared with that in the normal group.
The substantial elevation in caspase-3 and
caspase-9 protein expression was markedly
downregulated (p<0.001) by CoQ treat-
ment compared to the arsenic control
group. Treatment with rutin (50 and 100
mg/kg) significantly (p<0.01 and p<0.001)
and dose-dependently reduced both cas-
pase-3 and caspase-9 relative densities com-
pared to the arsenic control (Figs. 1A and
1B). Furthermore, Figs. 1C and 1D reveal
a strong inverse correlation between sperm
count and the relative densities of caspase-3
( = 0.8168, p<0.001) and Caspase-9 (R²
= 0.7075, p<0.01), respectively, indicating
that lower sperm counts are associated with
increased apoptotic activity.
Attenuation of arsenic-induced alteration
in testicular histopathology by rutin
Fig. 2 illustrates the histopathologi-
cal changes in rat testicular tissue after
exposure to arsenic and their ameliora-
tion across different treatment groups. In
the normal group (Fig. 2A), testicular ar-
chitecture appeared normal, with intact
seminiferous tubules containing organized
spermatogenic cells and healthy Leydig
cells, and no evidence of necrosis. However,
it showed mild inflammation (indicated by
black arrows). In contrast, the arsenic con-
Table 3. Attenuation of arsenic-induced alteration in testicular oxido-nitrosative stress
and inflammatory markers by rutin.
Parameters Normal As Control CoQ (10) R (25) R (50) R (100)
SOD (U/mg
of protein) 13.20 ± 1.10 5.19 ± 0.93### 10.66 ± 0.87*** 5.45 ± 0.61 8.91 ± 0.93** 10.66 ± 0.96***
GSH (µg/mg
of protein) 13.76 ± 0.64 3.90 ± 0.62### 11.04 ± 0.59*** 5.36 ± 0.50 8.31 ± 0.76** 10.43 ± 0.56***
MDA (nM/mg
of protein) 0.46 ± 0.10 3.14 ± 0.18### 1.97 ± 0.18*** 3.00 ± 0.27 2.11 ± 0.04** 1.59 ± 0.19***
NO (µg/mg
of protein) 119.00 ± 9.08 284.00 ± 13.96### 143.90 ± 9.97*** 261.40 ± 13.64 222.10 ± 12.14** 167.90 ± 9.82***
TNF-α (pg/mL) 157.00 ± 8.15 409.70 ± 12.69### 222.80 ± 14.07*** 377.20 ± 14.06 330.30 ± 8.02** 253.70 ± 12.06***
IL-1β (pg/mL) 17.28 ± 3.86 72.12 ± 1.85### 30.57 ± 2.76*** 68.46 ± 1.62 48.95 ± 2.70** 42.78 ± 2.41***
IL-6 (pg/mL) 40.50 ± 5.53 152.60 ± 5.18### 59.41 ± 9.32*** 155.50 ± 6.21 130.40 ± 7.87** 71.78 ± 7.13***
Data analysis: One-way ANOVA (post-hoc test: Tukey’s multiple range test). Data are reported as mean ± SEM
(n=6). Statistically significant compared with ###normal rats, ** and ***As control rats. ###p < 0.001, **p < 0.01
and ***p < 0.001. Arsenic (As), Co-enzyme Q10 (CoQ (10)), Glutathione (GSH), Interleukin-1 beta (IL-1β), Inter-
leukin-6 (IL-6), Malondialdehyde (MDA), Nitric Oxide (NO), Rutin (R), Superoxide Dismutase (SOD), and Tumor
Necrosis Factor-alpha (TNF).
416 Wang et al.
Investigación Clínica 66(4): 2025
trol group (Fig. 2B) demonstrated histo-
logical aberrations characterized by severe
inflammatory infiltration (black arrows),
necrosis (red arrows), vacuolated spermato-
genic cells (blue arrows), and Leydig cell
damage, indicating arsenic-induced toxic-
ity. The quantitative histopathological score
in Fig. 2G supports these visual observa-
tions, as the arsenic control group exhibited
significantly (p<0.001) elevated scores for
inflammatory infiltration, necrosis, Leydig
cell damage, and vacuolated spermatogenic
cells than the normal group. Treatment with
CoQ significantly reduced (p<0.001) these
histopathological aberrations compared to
the arsenic control group (Fig. 2C), sug-
gesting a protective effect against arsenic-
induced testicular injury. The rutin (25 mg/
kg) group showed a modest reduction in
these scores, but this was not significantly
Fig. 1. Attenuation of arsenic-induced alterations in testicular caspase-3 (A) and caspase-9 (B) protein ex-
pression by rutin. Correlation of sperm count with caspase-3 (C) and caspase-9 (D) protein expres-
sion. Data analysis: One-way ANOVA (post-hoc test: Tukey’s multiple range test). Data are reported as
mean ± SEM (n=6). Statistically significant compared with ###normal rats, ** and ***As control rats.
### p < 0.001, **p<0.01 and ***p<0.001. Correlation coefficients were determined using a two-sided
Fisher’s test. Arsenic (As), Co-enzyme Q10 (CoQ (10)), Glyceraldehyde-3-Phosphate Dehydrogenase
(GAPDH), and Rutin (R). Inflammatory infiltration (black arrow), necrosis (red arrow), and Leydig
cells damage (blue arrow).
Protective effects of rutin against arsenic-induced testicular damage 417
Vol. 66(4): 408 - 425, 2025
Fig. 2. Attenuation of arsenic-induced alterations in testicular histopathology by rutin treatment. Repre-
sentative images of testes from each group (A-F). Quantitative analysis of arsenic-induced alterations
in testicular histopathology and its attenuation by rutin (G). Data analysis: One-way ANOVA (post-hoc
test: Kruskal–Wallis test). Data are reported as mean ± SEM (n=6). Statistically significant compared
with ###normal rats, ** and ***As control rats. ###p < 0.001, **p < 0.01 and ***p < 0.001. Arsenic
(As), Co-enzyme Q10 (CoQ (10)), Glyceraldehyde-3-Phosphate Dehydrogenase (GAPDH), and Rutin (R).
418 Wang et al.
Investigación Clínica 66(4): 2025
different from the higher rutin or CoQ doses
(Fig. 2D). Treatment with rutin (50 and 100
mg/kg) showed a significant (p<0.01 and
p<0.001) reduction in arsenic-induced his-
tological aberrations in the testes, reflected
in a decrease in inflammatory infiltration,
necrosis, Leydig cell damage, and vacuolat-
ed spermatogenic cells compared to the ar-
senic control group (Figs 2E, 2F, and 2G).
DISCUSSION
Arsenic exposure has extensive human
health effects, with systemic toxicity affect-
ing multiple organs. It is particularly notori-
ous for its carcinogenic properties, causing
skin, lung, and bladder cancers, among oth-
ers 6. However, it is crucial to evaluate its ad-
verse effects on male reproductive health as
it directly affects male fertility. Arsenic expo-
sure impairs spermatogenesis and reduces
sperm quality through oxidative stress and
interference with crucial hormonal signaling
pathways 11. Researchers have explored the
protective efficacy of phytonutrients, such
as lutein and α-lipoic acid, against arsenic-
induced oxidative damage in testicular tis-
sues 36. In the present study, rutin, an herbal
intervention, was evaluated for its effects on
arsenite-induced testicular damage, and the
findings suggested that rutin ameliorated
testicular toxicity through its anti-apoptot-
ic, anti-inflammatory, and mitochondrial-
protective effects in experimental rats.
Integrative proteomic and metabolo-
mic analyses have shown that arsenic ex-
posure significantly alters the proteome
and metabolome in rat testes, affecting
spermatogenesis and fertilization through
disrupted signaling pathways 37. Sodium ar-
senite causes significant testicular damage
primarily through oxidative stress, apop-
tosis, and inflammatory pathways. Studies
have demonstrated that chronic exposure
to sodium arsenite can lead to a significant
decrease in both absolute and relative tes-
ticular weight 38. This reduction in testicu-
lar weight is attributed to the toxic effects
of arsenic on testicular tissue and its inter-
ference with normal spermatogenesis and
enzyme activities, including decreased acid
phosphatase, sorbitol dehydrogenase, and
17beta-hydroxy-steroid dehydrogenase activ-
ities, while increasing lactate dehydrogenase
and gamma-glutamyl transpeptidase activi-
ties. Additionally, arsenite exposure leads to
a notable increase in abnormal sperm forms,
along with a decrease in sperm count and
motility 38. Furthermore, arsenic is known to
accumulate significantly in reproductive tis-
sues, underscoring its potential to cause pro-
longed toxic effects 38. This accumulation in
the testes, epididymis, and prostate is linked
to oxidative stress and histopathological al-
terations, indicating that arsenic’s effects
are more profound at the cellular level 8,14.
In the present study, administration of ru-
tin significantly attenuated arsenite-induced
decreases in testes, epididymis, and prostate
weights, suggesting its protective efficacy in
reproductive organs.
Sodium arsenite led to decreased levels
of serum testosterone, luteinizing hormone,
and follicle-stimulating hormone, and to
significant changes in sperm parameters,
including reduced sperm count and motil-
ity, and an increase in abnormal sperm15.
Serum hormone levels, such as testosterone,
FSH, and LH, play crucial roles in regulating
male reproductive function 39. Testosterone,
which is primarily produced in the testes,
is vital for normal male reproductive func-
tions and secondary sexual characteristics.
FSH and LH, secreted by the pituitary gland,
regulate testicular function, including ste-
roidogenesis and spermatogenesis 40. Chron-
ic exposure to arsenite is often associated
with reduced testosterone levels owing to
impaired testicular function. This can be at-
tributed to the direct cytotoxic effects of ar-
senite on Leydig cells, which are responsible
for testosterone production. Additionally, al-
terations in hormone levels can disrupt the
hypothalamic-pituitary-gonadal axis, leading
to compensatory changes in LH and FSH lev-
els. In this study, arsenite-induced testicular
Protective effects of rutin against arsenic-induced testicular damage 419
Vol. 66(4): 408 - 425, 2025
damage was reflected in changes in serum
testosterone, FSH, and LH levels. However,
administration of rutin restored the dimin-
ished levels of these hormones in the serum.
Oxidative stress plays a crucial role in
arsenic-induced toxicity, impacting various
biological systems and leading to significant
health issues. Arsenic, a toxic metalloid, can
cross cellular barriers and accumulate in tis-
sues, contributing to oxidative stress by gen-
erating ROS 41,42. ROS generation is central
to the pathogenesis of arsenic toxicity, as it
leads to mitochondrial dysfunction, a criti-
cal factor in neurotoxicity, and other health
issues 43,44. Arsenic-induced oxidative stress
leads to mitochondrial damage by disrupting
the normal balance between antioxidants
and pro-oxidants within cells. This disruption
impairs mitochondrial membrane potential,
promotes cytochrome c release, and triggers
apoptosis through caspase activation42. On
a systemic level, oxidative stress also causes
chromosomal instability and DNA damage,
leading to further complications, such as car-
cinogenesis and neurological disorders44-47.
Moreover, arsenic exposure has been shown
to deplete antioxidant capacities and elevate
oxidative damage biomarkers, such as malo-
ndialdehyde (MDA) and nitric oxide, owing
to the heightened oxidative environment48,49.
These changes underline the oxidative stress
mechanism as a cornerstone of arsenic tox-
icity, which affects various systems, includ-
ing the reproductive system 50. Therapeutic
strategies to combat arsenic-induced oxida-
tive stress focus on enhancing mitochondrial
function and increasing antioxidant defense.
Acetyl-L-carnitine (ALC), for instance, has
been shown to counteract arsenic-induced
oxidative stress by improving antioxidant
mechanisms and mitochondrial function,
thus offering a potential therapeutic pathway
42. Other antioxidants, such as curcumin and
apigenin, also exhibit protective effects by
reducing ROS and supporting cellular anti-
oxidant systems, highlighting their possible
roles in mitigating arsenic toxicity51,52. In the
present investigation, arsenic-induced toxic-
ity, associated with elevated oxidative stress,
contributed to various cellular and systemic
damage, as reflected by diminished testicu-
lar mitochondrial enzyme activity. However,
rutin treatment effectively increased tes-
ticular glutathione and superoxide levels
and reduced malondialdehyde and nitric ox-
ide levels, suggesting its protective effects
against arsenic-induced mitochondrial dys-
function, potentially by mitigating oxidative
stress and enhancing mitochondrial energy
production.
Inflammation is a significant contribu-
tor to arsenic-induced testicular toxicity.
Pro-inflammatory cytokines, such as TNF-α
and interleukins, mediate this process.
TNF-α induces apoptosis and promotes in-
flammatory responses 53. Inflammation
induced by TNF-α can activate various sig-
naling pathways, culminating in oxidative
stress and tissue damage 54-56. It has been ob-
served that arsenic exposure results in the
upregulation of inflammatory mediators,
such as TNF-α and interleukins, contribut-
ing to testicular damage 57. Furthermore,
Caspases 3 and 9 play crucial roles in the
apoptotic pathways underlying arsenic-in-
duced testicular toxicity. Caspase-9 is part
of the intrinsic or mitochondrial apoptosis
pathway, which is often activated by cellular
stressors, including toxins such as arsenic.
It is responsible for activating downstream
effector caspases, including caspase-3 58,59.
Caspase-3 then executes apoptosis by cleav-
ing cellular substrates, leading to system-
atic breakdown of cellular components and
cell death 60-62. In arsenic-induced testicu-
lar toxicity, activation of these caspases is
indicative of enhanced apoptotic activity,
contributing to the observed tissue damage
and dysfunction 57. In the current study,
arsenic-induced elevated inflammation and
apoptosis, mediated by cytokines such as
TNF-α and enzymes such as caspases 3 and
9, highlighted the development of arsenic-
induced testicular toxicity. However, rutin
treatment mitigated the toxic effects in-
duced by chronic exposure to arsenic via
420 Wang et al.
Investigación Clínica 66(4): 2025
the amelioration of these pathways, sug-
gesting its therapeutic potential against
arsenic-induced testicular toxicity.
This study has some limitations. First,
the study focused on acute arsenic exposure
(two consecutive days), which may not re-
flect the effects of chronic, long-term expo-
sure often observed in human populations.
Second, the study only used sodium arsenite,
whereas environmental arsenic exposure of-
ten involves multiple arsenic species. Third,
this study did not address the bioavailabil-
ity and metabolism of rutin in rats, which
could affect its efficacy in humans. Lastly,
the study focused on rutin alone and did not
explore its potential synergistic effects with
other protective agents.
As a conclusion, rutin demonstrated
significant protective effects against sodium
arsenite-induced testicular toxicity in rats.
Rutin ameliorated arsenic-induced damage
by reducing oxidative stress, inflammation,
and apoptosis in testicular tissue, while im-
proving sperm parameters and hormone lev-
els. These findings suggest that rutin may
have therapeutic potential in mitigating ar-
senic-related reproductive toxicity, although
further research is needed to elucidate its
mechanisms of action and clinical applica-
bility fully.
Acknowledgement
Medical writing support for the devel-
opment of this manuscript, under the direc-
tion of the authors, was provided by Yonnova
Scientific Consultancy Pvt. Ltd. in accor-
dance with the Good Publication Practice
guidelines.
Funding
This study was funded by the Zheji-
ang Province Medical and Health Science
and Technology Plan Project (2021RC096)
and the Zhejiang Traditional Chinese Medi-
cine Science and Technology Plan Project
(2023ZL370).
Conflicts of interest
The authors declare that they have no
conflict of interest regarding this study.
Availability of data
The datasets generated and/or analyzed
during the current study are not publicly
available due to confidentiality agreements;
supporting data can only be made available
to bona fide researcher’s subject to a non-
disclosure agreement.
Ethical approval
All experiments were performed be-
tween 09:00 and 17:00. The experimental
protocol was approved by the Institutional
Animal Ethics Committee of the Shaanxi
Kangfu Hospital (approval no. sxkf2025).
All procedures involving animals were con-
ducted in accordance with the National In-
stitute of Health Guide for the Care and Use
of Laboratory Animals.
ORCID number of authors
Yunyun Wang (YW):
0009-0007-3462-5323
Zengrong Hua (ZH):
0009-0008-8248-8148
Jin Qing Hui (JQH):
0009-0008-5583-9213
Dandan Qiu (DQ):
0009-0009-1376-7853
Author contributions
Each author has made significant con-
tributions to the development of this manu-
script. YW: conceived and designed the eval-
uation and drafted the manuscript; ZH and
DQ: performed data acquisition; YW: par-
ticipated in developing the assessment, per-
formed parts of the statistical analysis, and
helped to draft the manuscript; JQH: revised
the manuscript and performed the statisti-
cal analysis. All the authors have read and
approved the final version of the manuscript.
Protective effects of rutin against arsenic-induced testicular damage 421
Vol. 66(4): 408 - 425, 2025
REFERENCES
1. Adil M, Kandhare AD, Ghosh P, Bod-
hankar SL. Sodium arsenite-induced myo-
cardial bruise in rats: Ameliorative effect
of naringin via TGF-β/Smad and Nrf/HO
pathways. Chem Biol Interact. 2016;253:
66-77. https://doi.org/10.1016/j.cbi.2016.
05.015.
2. Adil M, Kandhare AD, Visnagri A, Bod-
hankar SL. Naringin ameliorates sodium
arsenite-induced renal and hepatic toxi-
city in rats: decisive role of KIM-1, Caspa-
se-3, TGF-b, and TNF-a. Ren Fail. 2015;37
(8):1396-1407. https://doi.org/10.3109/0
886022X.2015.1074462
3. Kim YJ, Kim JM. Arsenic Toxicity in Male
Reproduction and Development. Dev Re-
prod. 2015;19 (4):167-180. https://doi.
org/10.12717/DR.2015.19.4.167.
4. Rahmani S, Naraki K, Roohbakhsh A, Ha-
yes AW, Karimi G. The protective effects
of rutin on the liver, kidneys, and heart
by counteracting organ toxicity caused by
synthetic and natural compounds. Food
Sci Nutr. 2022;11 (1):39-56. https://doi.
org/10.1002/fsn3.3041.
5. World Health Organization. Obesity and
overweight [Internet]. Geneva: World
Health Organization; 2020. Available from:
https://www.who.int/news-room/fact-
sheets/detail/obesity-and-overweight
6. Hong YS, Song KH, Chung JY. Health
effects of chronic arsenic exposure. J Prev
Med Public Health. 2014;47(5):245-252.
https://doi.org/10.3961/jpmph.14.035.
7. Mukherjee AA, Kandhare AD, Bodhankar
SL. Elucidation of protective efficacy of
Pentahydroxy flavone isolated from Mad-
huca indica against arsenite-induced
cardiomyopathy: Role of Nrf-2, PPAR-
gamma, c-fos and c-jun. Environ Toxicol
Pharmacol. 2017;56:172-185. https://doi.
org/10.1016/j.etap.2017.08.027
8. Souza AC, Marchesi SC, Ferraz RP, Lima
GD, de Oliveira JA, Machado-Neves M.
Effects of sodium arsenate and arseni-
te on male reproductive functions in
Wistar rats. J Toxicol Environ Health A.
2016;79(6):274-286. https://doi.org/10.1
080/15287394.2016.1150926.
9. Zubair M, Ahmad M, Qureshi ZI. Review
on arsenic-induced toxicity in male repro-
ductive system and its amelioration. An-
drologia. 2017; 49(9):e12791. https://doi.
org/10.1111/and.12791.
10. Rachamalla M, Chinthada J, Kushwa-
ha S, Putnala SK, Sahu C, Jena G, et al.
Contemporary Comprehensive Review on
Arsenic-Induced Male Reproductive Toxici-
ty and Mechanisms of Phytonutrient Inter-
vention. Toxics. 2022; 10(12):744. https://
doi.org/10.3390/toxics10120744.
11. Souza ACF, Bastos DSS, Sertorio MN,
Santos FC, Ervilha LOG, de Oliveira LL,
et al. Combined effects of arsenic expo-
sure and diabetes on male reproductive
functions. Andrology. 2019; 7(5):730-740.
https://doi.org/10.1111/andr.12613.
12. Wang S, Zhao Y, Wu L, Tang M, Su C, Hei
TK, Yu Z. Induction of germline cell cycle
arrest and apoptosis by sodium arsenite
in Caenorhabditis elegans. Chem Res To-
xicol. 2007;20(2):181-186. https://doi.
org/10.1021/tx0601962.
13. Silva CS, Kudlyk T, Tryndyak VP, Twaddle
NC, Robinson B, Gu Q, et al. Gene expres-
sion analyses reveal potential mechanism of
inorganic arsenic-induced apoptosis in ze-
brafish. J Appl Toxicol. 2023;43(12):1872-
1882. https://doi.org/10.1002/jat.4520.
14. Uygur R, Aktas C, Caglar V, Uygur E, Er-
dogan H, Ozen OA. Protective effects of
melatonin against arsenic-induced apopto-
sis and oxidative stress in rat testes. Toxicol
Ind Health. 2016;32(5):848-859. https://
doi.org/10.1177/0748233713512891.
15. Omar SM, Zahran NN, Alhotan RA, Hus-
sein EO, Galik B, Saleh AA. Evaluation of
Salvia hispanica as a Therapeutic Agent
against Sodium Arsenic-Induced Testicular
Toxicity in a Male Rats Model. Life (Basel
Switzerland). 2024; 14(1):109. https://
doi.org/10.3390/life14010109.
16. Pires Das Neves RN, Carvalho F, Carval-
ho M, Fernandes E, Soares E, de Bas-
tos ML, et al. Protective activity of hes-
peridin and lipoic acid against sodium
arsenite acute toxicity in mice. Toxicol
Pathol. 2004;32(5):527-535. https://doi.
org/10.1080/01926230490502566.
422 Wang et al.
Investigación Clínica 66(4): 2025
17. Negahdari R, Bohlouli S, Sharifi S, Male-
ki Dizaj S, Rahbar Saadat Y, Khezri K, et
al. Therapeutic benefits of rutin and its
nanoformulations. Phytother Res. 2021;
35(4):1719-1738. https://doi.org/10.100
2/ptr.6904.
18. Budzynska B, Faggio C, Kruk-Slomka M,
Samec D, Nabavi SF, Sureda A, et al. Ru-
tin as Neuroprotective Agent: From Bench
to Bedside. Curr Med Chem. 2019;26
(27):5152-5164. https://doi.org/10.2174/
0929867324666171003114154.
19. Hosseinzadeh H, Nassiri-Asl M. Review of
the protective effects of rutin on the me-
tabolic function as an important dietary
flavonoid. J Endocrinol Invest. 2014;37
(9):783-788. https://doi.org/10.1007/s4
0618-014-0096-3.
20. Ganeshpurkar A, Saluja AK. The Pharma-
cological Potential of Rutin. Saudi Pharm
J. 2017;25 (2):149-164. https://doi.org/
10.1016/j.jsps.2016.04.025
21. Niture NT, Ansari AA, Naik SR. Anti-hy-
perglycemic activity of rutin in strepto-
zotocin-induced diabetic rats: an effect
mediated through cytokines, antioxidants
and lipid biomarkers. Indian J Exp Biol.
2014;52 (7):720-727. PMID: 25059040.
22. Kamalakkannan N, Prince PS. Antihyper-
glycaemic and antioxidant effect of rutin,
a polyphenolic flavonoid, in streptozo-
tocin-induced diabetic wistar rats. Basic
Clin Pharmacol Toxicol. 2006;98:97-103.
https://doi.org/10.1111/j.1742-7843.2006.
pto_241.x
23. Das J, Ghosh J, Manna P, Sinha M, Sil PC.
Taurine protects rat testes against NaAsO2
induced oxidative stress and apoptosis via
mitochondrial dependent and independent
pathways. Toxicol Lett. 2009;187(3):201-
210. https://doi.org/10.1016/j.toxlet.200
9.03.001.
24. Adil M, Visnagri A, Kumar VS, Kandhare
AD, Ghosh P, Bodhankar SL. Protective
effect of naringin on sodium arsenite in-
duced testicular toxicity via modulation of
biochemical perturbations in experimen-
tal rats. Pharmacologia. 2014;5 (6):222-
234. https://doi.org/10.5567/pharmaco-
logia.2014.222.234
25. Thangapandiyan S, Miltonprabu S. Epi-
gallocatechin gallate exacerbates fluo-
ride-induced oxidative stress mediated
testicular toxicity in rats through the
activation of Nrf2 signaling pathway.
Asian Pac J Reprod. 2015;4 (4):272-287.
https://doi.org/https://doi.org/10.1016/j.
apjr.2015.07.005
26. Kandhare AD, Kumar VS, Adil M, Rajmane
AR, Ghosh P, Bodhankar SL. Investigation
of gastro protective activity of Xanthium
strumarium L. by modulation of cellular
and biochemical marker. Orient Pharm
Exp Med. 2012;12: 287-299. https://doi.
org/10.1007/s13596-012-0090-2
27. Kandhare AD, Raygude KS, Ghosh P,
Ghule AE, Bodhankar SL. Neuroprotec-
tive effect of naringin by modulation of
endogenous biomarkers in streptozotocin
induced painful diabetic neuropathy. Fito-
terapia. 2012;83 (4):650-659. https://doi.
org/10.1016/j.fitote.2012.01.010
28. Kandhare AD, Raygude KS, Ghosh P,
Ghule AE, Bodhankar SL. Therapeutic
role of curcumin in prevention of bioche-
mical and behavioral aberration induced
by alcoholic neuropathy in laboratory ani-
mals. Neurosci Lett. 2012;511(1):18-22.
https://doi.org/10.1016/j.neulet.2012.
01.019
29. Visnagri A, Kandhare AD, Shiva Kumar
V, Rajmane AR, Mohammad A, Ghosh P
et al. Elucidation of ameliorative effect of
Co-enzyme Q10 in streptozotocin-induced
diabetic neuropathic perturbation by mo-
dulation of electrophysiological, bioche-
mical and behavioral markers. Biomed
Aging Pathol. 2012;2 (4):157-172. https://
doi.org/http://dx.doi.org/10.1016/j.bio-
mag.2012.10.006
30. Adil M, Kandhare AD, Dalvi G, Ghosh P,
Venkata S, Raygude KS, Bodhankar SL.
Ameliorative effect of berberine against
gentamicin-induced nephrotoxicity in rats
via attenuation of oxidative stress, inflam-
mation, apoptosis and mitochondrial dys-
function. Ren Fail. 2016;38 (6):996-1006.
https://doi.org/10.3109/0886022X.201
6.1165120
Protective effects of rutin against arsenic-induced testicular damage 423
Vol. 66(4): 408 - 425, 2025
31. Ketkar S, Rathore A, Kandhare AD, Lohi-
dasan S, Bodhankar SL, Paradkar A, et
al. Alleviating exercise-induced muscular
stress using neat and processed bee pollen:
oxidative markers, mitochondrial enzy-
mes, and myostatin expression in rats. In-
tegr Med Res. 2015;4 (3):147-160. https://
doi.org/10.1016/j.imr.2015.02.003
32. Liu J, Han XC, Wang D, Kandhare AD,
Mukherjee-Kandhare AA, Bodhankar SL
et al. Elucidation of Molecular Mechanism
Involved in Nephroprotective Potential of
Naringin in Ethylene Glycol-Induced Uro-
lithiasis in Experimental Uninephrectomi-
zed Hypertensive Rats. Lat Am J Pharm.
2020;39 (5):991-999.
33. Yao X, Li L, Kandhare AD, Mukher-
jeeKandhare AA, Bodhankar SL. Atte-
nuation of reserpineinduced fibromyalgia
via ROS and serotonergic pathway modula-
tion by fisetin, a plant flavonoid polyphenol.
Exp Ther Med. 2020;19 (2):1343-1355.
https://doi.org/10.3892/etm.2019.8328
34. Zhang K, Kandhare A, Mukherjee-Kand-
hare A, Bodhankar S. Apigenin attenua-
ted ethylene glycol induced urolithiasis
in uninephrectomized hypertensive rats:
A possible role of bikunin, BMP-2/4, and
osteopontin. Pharmacogn Mag. 2020;16
(71):455-463. https://doi.org/10.4103/
pm.pm_83_20
35. McNamara ER, Madden-Fuentes RJ,
Routh JC, Rouse D, Madden JF, Wiener
JS, et al. Evaluation of cold ischemia for
preservation of testicular function during
partial orchiectomy in the rat model. J Pe-
diatr Urol. 2014;10 (4):593-597. https://
doi.org/10.1016/j.jpurol.2014.06.005
36. Prathima P, Pavani R, Sukeerthi S, Sai-
nath SB. α-Lipoic acid inhibits testicular
and epididymal oxidative damage and im-
proves fertility efficacy in arsenic-intoxica-
ted rats. J Biochem Mol Toxicol. 2018;32
(2):e22016. https://doi.org/10.1002/jbt.
22016.
37. Huang Q, Luo L, Alamdar A, Zhang J, Liu
L, Tian M, et al. Integrated proteomics
and metabolomics analysis of rat testis:
Mechanism of arsenic-induced male repro-
ductive toxicity. Sci Rep. 2016;6:32518.
https://doi.org/10.1038/srep32518.
38. Pant N, Murthy RC, Srivastava SP. Male
reproductive toxicity of sodium arse-
nite in mice. Hum Exp Toxicol. 2004;
23(8):399-403. https://doi.org/10.1191/
0960327104ht467oa.
39. Oduwole OO, Huhtaniemi IT, Misrahi M.
The Roles of Luteinizing Hormone, Follicle-
Stimulating Hormone and Testosterone in
Spermatogenesis and Folliculogenesis Re-
visited. Int J Mol Sci. 2021;22 (23):12735.
https://doi.org/10.3390/ijms222312735
40. Oduwole OO, Peltoketo H, Huhtaniemi
IT. Role of Follicle-Stimulating Hormo-
ne in Spermatogenesis. Front Endocri-
nol. (Lausanne). 2018;9:763. https://doi.
org/10.3389/fendo.2018.00763
41. Prakash C, Soni M, Kumar V. Mitochon-
drial oxidative stress and dysfunction in
arsenic neurotoxicity: A review. J Appl To-
xicol. 2016; 36 (2):179-188. https://doi.
org/10.1002/jat.3256
42. Keshavarz-Bahaghighat H, Sepand MR,
Ghahremani MH, Aghsami M, Sanadgol
N, Omidi A, et al. Acetyl-L-Carnitine At-
tenuates Arsenic-Induced Oxidative Stress
and Hippocampal Mitochondrial Dys-
function. Biol Trace Elem Res. 2018;184
(2):422-435. https://doi.org/10.1007/s1
2011-017-1210-0
43. Thakur M, Rachamalla M, Niyogi S, Da-
tusalia AK, Flora SJS. Molecular Mecha-
nism of Arsenic-Induced Neurotoxicity
including Neuronal Dysfunctions. Int J
Mol Sci. 2021;22 (18):10077. https://doi.
org/10.3390/ijms221810077
44. Liu L, Trimarchi JR, Navarro P, Blasco
MA, Keefe DL. Oxidative Stress Contribu-
tes to Arsenic-induced Telomere Attrition,
Chromosome Instability, and Apoptosis. J
Biol Chem. 2003;278 (34):31998-32004.
https://doi.org/10.1074/jbc.m303553200
45. Cui J, Wang G, Kandhare AD, Mukher-
jee-Kandhare AA, Bodhankar SL. Neu-
roprotective effect of naringin, a flavone
glycoside in quinolinic acid-induced neu-
rotoxicity: Possible role of PPAR-gamma,
Bax/Bcl-2, and caspase-3. Food Chem
424 Wang et al.
Investigación Clínica 66(4): 2025
Toxicol. 2018;121:95-108. https://doi.
org/10.1016/j.fct.2018.08.028
46. Kandhare AD, Mukherjee AA, Bodhankar
SL. Neuroprotective effect of Azadirachta
indica standardized extract in partial scia-
tic nerve injury in rats: Evidence from
anti-inflammatory, antioxidant and anti-
apoptotic studies. EXCLI J. 2017;16:546-
565. https://doi.org/doi: 10.17179/excli
2017-161.
47. Raygude KS, Kandhare AD, Ghosh P,
Ghule AE, Bodhankar SL. Evaluation of
ameliorative effect of quercetin in expe-
rimental model of alcoholic neuropathy
in rats. Inflammopharmacoly. 2012; 20
(6):331-341. https://doi.org/10.1007/s10
787-012-0122-z
48. Xu M, Rui D, Yan Y, Xu S, Niu Q, Feng
G, et al. Oxidative Damage Induced by
Arsenic in Mice or Rats: A Systematic Re-
view and Meta-Analysis. Biol Trace Elem
Res. 2017; 76 (1):154-175. https://doi.
org/10.1007/s12011-016-0810-4
49. Visnagri A, Kandhare AD, Chakravarty
S, Ghosh P, Bodhankar SL. Hesperidin,
a flavanoglycone attenuates experimental
diabetic neuropathy via modulation of ce-
llular and biochemical marker to impro-
ve nerve functions. Pharm Biol. 2014;52
(7):814-828. https://doi.org/10.3109/138
80209.2013.870584
50. Jomova K, Jenisova Z, Feszterova M, Ba-
ros S, Liska J, Hudecova D, et al. Arsenic:
toxicity, oxidative stress and human disea-
se. J Appl Toxicol. 2011;31 (2):95-107.
https://doi.org/10.1002/jat.1649
51. Biswas J, Sinha D, Mukherjee S, Roy S,
Siddiqi M, Roy M. Curcumin protects DNA
damage in a chronically arsenic-exposed
population of West Bengal. Hum Exp To-
xicol. 2010;29 (6):513-524. https://doi.
org/10.1177/0960327109359020
52. Almeer R, Alyami NM. The protective
effect of apigenin against inorganic ar-
senic salt-induced toxicity in PC12 cells.
Environ Sci Poll Res. 2023;30:106625-
106635. https://doi.org/10.1007/s11356-
023-29884-w
53. El-Khadragy M, Al-Megrin WA, Alsadhan
NA, Metwally DM, El-Hennamy RE, Sa-
lem FEH, et al. Impact of Coenzyme Q10
Administration on Lead Acetate-Induced
Testicular Damage in Rats. Oxid Med Cell
Longev. 2020;2020: 4981386. https://doi.
org/10.1155/2020/4981386
54. Devkar ST, Kandhare AD, Zanwar AA,
Jagtap SD, Katyare SS, Bodhankar SL, et
al. Hepatoprotective effect of withanolide-
rich fraction in acetaminophen-intoxica-
ted rat: decisive role of TNF-α, IL-1β, COX-
II and iNOS. Pharm Biol. 2016; 54 (11):
2394-2403. https://doi.org/10.3109/1388
0209.2016.1157193.
55. Honmore VS, Kandhare AD, Kadam PP,
Khedkar VM, Natu AD, Rojatkar SR, et
al. Diarylheptanoid, a constituent isolated
from methanol extract of Alpinia officina-
rum attenuates TNF-alpha level in Freund’s
complete adjuvant-induced arthritis in rats.
J Ethnopharmacol. 2019;229:233-245.
https://doi.org/10.1016/j.jep.2018.10.019
56. Kandhare AD, Bodhankar SL, Mohan V,
Thakurdesai PA. Effect of glycosides ba-
sed standardized fenugreek seed extract
in bleomycin-induced pulmonary fibrosis
in rats: Decisive role of Bax, Nrf2, NFB,
Muc5ac, TNF-α and IL-1β. Chem Biol In-
teract. 2015; 237:151-165. https://doi.
org/10.1016/j.cbi.2015.06.019.
57. Rao G, Qiao B, Zhong G, Li T, Su Q, Wu
S, et al. Arsenic and polystyrene-nano plas-
tics co-exposure induced testicular toxici-
ty: Triggers oxidative stress and promotes
apoptosis and inflammation in mice. Envi-
ron Toxicol. 2023;39 (1):264-276. https://
doi.org/10.1002/tox.23970
58. Joza N, Susin SA, Daugas E, Stanford
WL, Cho SK, Li CY, et al. Essential role
of the mitochondrial apoptosis-inducing
factor in programmed cell death. Natu-
re 2001;410(6828):549-554. https://doi.
org/10.1038/35069004
59. Zhao P, Wang Y, Yang Q, Yu G, Ma F, Dong
J. Abamectin causes cardiac dysfunction in
carp via inhibiting redox equilibrium and
resulting in immune inflammatory respon-
se and programmed cell death. Environ Sci
Pollut Res Int. 2023;30 (11):29494-29509.
https://doi.org/10.1007/s11356-022-
24004-6
Protective effects of rutin against arsenic-induced testicular damage 425
Vol. 66(4): 408 - 425, 2025
60. Ma T, Kandhare AD, Mukherjee-Kandhare
AA, Bodhankar SL. Fisetin, a plant fla-
vonoid ameliorates doxorubicin-induced
cardiotoxicity in experimental rats: the
decisive role of caspase-3, COX-II, cTn-I,
iNOs and TNF-alpha. Mol Biol Rep 2019;46
(1):105-118. https://doi.org/10.1007/
s11033-018-4450-y
61. Visnagri A, Kandhare AD, Bodhankar SL.
Renoprotective effect of berberine via in-
tonation on apoptosis and mitochondrial-
dependent pathway in renal ischemia
reperfusion-induced mutilation. Ren Fail.
2015;37(3):482-493. https://doi.org/10.3
109/0886022X.2014.996843
62. Ghule AE, Kandhare AD, Jadhav SS, Zan-
war AA, Bodhankar SL. Omega-3-fatty
acid adds to the protective effect of flax
lignan concentrate in pressure overload-
induced myocardial hypertrophy in rats via
modulation of oxidative stress and apop-
tosis. Int Immunopharmacol. 2015; 28
(1):751-763. https://doi.org/10.1016/j.
intimp.2015.08.005.