Abstract
Background
Medications, including chemotherapeutic drugs, contribute to male infertility as external factors by inducing oxidative stress in testicular cells. Shilajit is a naturally occurring bioactive antioxidant used in Ayurvedic medicine to treat a variety of ailments.
Objective
This study examines the potential of Shilajit to counteract the negative effects of the chemotherapeutic drug cyclophosphamide (CPA) on testicular germ cell dynamics.
Material and methods
Male Parkes mice received single intraperitoneal CPA injection (200 mg/kg BW) on day one, followed by daily supplementation of Shilajit (100 and 200 mg/kg BW) for one spermatogenic cycle.
Results
CPA adversely affected testicular germ cell dynamics by inhibiting the conversion of spermatogonia-to-spermatids, altering testicular histoarchitecture, impairing Sertoli cell function and testicular steroidogenesis, and disturbing the testicular oxido-apoptotic balance. Shilajit supplementation restores testicular germ cell dynamics in CPA-exposed mice, as evidenced by improved histoarchitecture of the testis. Shilajit improves testicular daily production and sperm quality by promoting the conversion of spermatogonia (2C) into spermatids (1C), stimulating germ cell proliferation (PCNA), improving Sertoli cell function (N-Cadherin and β-Catenin), and maintaining the Bax/Bcl2 ratio. Additionally, Shilajit enhances testosterone biosynthesis by activating enzymes like 3β-HSD, and 17β-HSD. Shilajit also reduces testicular oxidative stress by increasing antioxidant enzyme activity (SOD) and decreasing lipid peroxidation (LPO). These effects are mediated by upregulation of the antioxidant protein Nrf-2 and downregulation of Keap-1.
Conclusion
The findings underscore the potent androgenic and antioxidant characteristics of Shilajit, as well as its ability to enhance fertility in cases of testicular damage caused by chemotherapeutic drugs.
Keywords: Cyclophosphamide, Shilajit, Oxidative stress, Steroidogenesis, Testicular germ cell dynamics, Sertoli cell function
Highlights
-
•
CPA impairs steroidogenesis and germ cell dynamics by inducing oxidative stress.
-
•
Shilajit restore steroidogenesis and Sertoli cell function in CPA-exposed mice.
-
•
Shilajit ameliorates daily sperm production and sperm quality in CPA-exposed mice.
-
•
Shilajit restores germ cell dynamics by restoring 1C:2C ratio in CPA-exposed mice.
-
•
Shilajit regulates oxidative stress via Nrf-2/Keap-1 signaling in CPA-exposed mice.
1. Introduction
Sexual health is essential for overall physical and emotional well-being [1]. Over the past three decades, male infertility has emerged as a significant global health concern, affecting approximately 12–15% of couples worldwide. Notably, male factors contribute to 50% of all infertility cases [2,3]. Among the various factors responsible for the rising incidence of male infertility, the use of medications has been identified as a common external contributor to male infertility [2,4]. Chemotherapy drugs, which have played a significant role in the recent increase in cancer patient survival rates, are designed to target and kill rapidly dividing cancer cells; however, they can also affect other rapidly dividing cells in the body, such as proliferating germ cells in the testes [5,6]. Chemotherapeutic drugs cause excessive oxidative stress [7,8], which appears to have a negative impact on spermatogenesis, testicular steroidogenesis and may also impair Sertoli cell function related to the maintenance of blood-testis barriers [9,10]. This can lead to long-term or permanent male infertility, which can lead to depression, mood swings, and psychological instability in cancer survivors, preventing them from having a normal family life [11,12]. Therefore, shielding the testicular germ cell dynamics from the side effects of cancer treatments may be a promising way to preserve male fertility while undergoing cancer treatment. Recent research suggests that supplementing with antioxidants may be a viable approach to reducing the toxicity caused by chemotherapeutic drugs [13]. Furthermore, naturally occurring antioxidant elements have been shown to be more effective than synthetic antioxidants in mitigating a variety of oxidative stress-related conditions, including male reproductive health disorders [14,15].
Shilajit is a blackish-brown herbomineral natural medicine collected from high-altitude Himalayan rocks of the Indian subcontinent [16,17]. Its chemical composition has been well characterized, and it has been discovered to contain a variety of beneficial components. These include Fulvic Acid and Humic Acid, which are said to be responsible for its health benefits. Shilajit also contains Dibenzo--Pyrones (DBPs), which have antioxidant properties. It also contains amino acids, tannins, phytosterols, and terpenoids, as well as over 40 trace minerals [[18], [19], [20]]. Shilajit has been shown in current scientific studies to have variety of pharmacological properties, such as antioxidant, antidiabetic, anti-aging, rejuvenating, and anti-inflammatory [18,21,22]. In addition, current scientific research also points the role of Shilajit in the prevention of cancer [23,24]. Shilajit is an important revitalising agent in the ancient Indian healing system of Ayurveda. Traditional practitioners in the Indian subcontinent have used its properties to address male sexual health issues [25]. Some scientific studies also support the beneficial effects of Shilajit on male reproductive health [18,21]. Furthermore, our earlier investigations revealed that Shilajit has the potential to safeguard male reproductive health from heavy metal exposure [25,26]. Despite being renowned for its antioxidant, rejuvenating, and anti-cancer properties, there has been a lack of comprehensive studies examining the potential of utilizing Shilajit as a natural antioxidant to mitigate the side effects of chemotherapeutic drugs on testicular germ cell dynamics.
The objective of this current study was to explore the potential of Shilajit in safeguarding testicular germ cell dynamics from the side effects of chemotherapeutic drugs. We specifically chose cyclophosphamide (CPA), a widely recognized chemotherapeutic agent with established antitumor and immunosuppressive properties [27,28]. CPA is used to treat acute and chronic leukaemias, multiple myeloma, lymphoma, rheumatoid arthritis, and ovarian and breast cancer [29,30]. However, due to its nonspecific action, it affects rapidly dividing testicular cells by inducing excessive oxidative stress, thereby leading to infertility in cancer survivors [31,32]. This underscores the need to explore the potential of a well-known natural antioxidant and rejuvenator, Shilajit, in preventing these detrimental effects. Therefore, we employed various techniques, including histology, sperm analysis, immunoblot analysis, flow cytometry, biochemical estimations, and ELISA, to evaluate the efficacy of Shilajit in maintaining testicular germ cell dynamics in male mice exposed to cyclophosphamide. The objective was to establish Shilajit as a natural remedy capable of shielding the testes from the side effects associated with anticancer medications.
2. Materials and methods
2.1. Chemicals
In the present investigation, unless otherwise indicated, chemicals were purchased from Sigma, Merck India Ltd. and SRL Pvt. Ltd, India.
2.2. Test materials and preparation of dose
Purified Shilajit (Shilajit-964; Batch No.:20201001) was obtained from VCA Healthcare Pvt Limited (G-91 Agro Food Park, MIA, Alwar, Rajasthan, India), with percentages of total fulvic acid and dibenzopyrones of 40% and 5%, respectively (according to the manufacturer's certificate of analysis). Shilajit (100 or 200 mg/kg body weight) was dissolved in double distilled water and given orally for one spermatogenic cycle (35 days). Cyclophosphamide (Endoxan; Batch No.: BUX1023) was purchased from Baxter Oncology GMBH (Westfalen, Germany). The drug was dissolved in normal saline, and a single intraperitoneal (ip) dose of cyclophosphamide at a concentration of 200 mg/kg of body weight was administered to mice on the first day of the experiment. The doses of cyclophosphamide were chosen based on Oyagbemi et al. [14]while the Shilajit doses were chosen based on our previous studies [25,26].
2.3. Animal care, maintenance and experimental design
Forty Parkes (P) strain adult male mice (14–15 weeks old) weighing 36g–42 g were used for the current study. The mice were kept in clean and hygienic conditions within a well-ventilated room, maintaining a temperature of 25 ± 2°C and a 12-h light-dark cycle. The relative humidity was maintained at 50 ± 10%. The mice were provided with pellet meal (VRK Nutritional Solutions, Laboratory Animal Feeds, Maharashtra, India) and access to water as required. Animals of each experimental group were housed in a different set of polypropylene cages (29 cm × 22 cm x 14 cm) with dry rice husk used as a bedding material. The Animal Ethics Committee (BHU/DoZ/IAEC/2020/02/035) of Banaras Hindu University approved the experimental design. Mice were randomly allocated to one of four experimental groups (10 mice/group) as follows:
-
1.
Control: Vehicle control (200μl DDW/oral/day for 35 Days)
-
2.
CPA: Cyclophosphamide (200 mg/kg BW, single ip injection on first day of experiment)
-
3.
CPA + S100: Cyclophosphamide (200 mg/kg BW, single ip injection on first day of experiment) followed by Shilajit treatment (100 mg/kg BW/day) for 35 days
-
4.
CPA + S200: Cyclophosphamide (200 mg/kg BW, single ip injection on first day of experiment) followed by Shilajit treatment (200 mg/kg BW/day) for 35 days
2.4. Sample collection
The mice were euthanized 24 h following the final administration of Shilajit. At autopsy, coronary blood samples were collected after recording the body weight of each animal. The blood samples were then processed to obtain serum which was preserved at −80 °C for later analysis. The testes were surgically excised, carefully cleaned of any residual blood precisely and weighed to an accuracy of 0.10mg. Histology and immunohistochemical localization were performed on one testis from each of five different mice. The contralateral testis of five mice were utilized to evaluate oxidative stress markers and to perform immunoblot analysis. Remaining five testes from five different mice were employed to assess daily sperm production. Contralateral testis from five different mice was used for flow cytometry analysis (see Supplementary Fig. 1). Cauda epididymidis sperm analysis was performed on six animals.
2.5. Sperm analysis (count, motility, viability and morphology)
Preparation of Sperm Suspensions: Sperm suspensions were prepared by mincing the caudal epididymis in 0.5ml of normal saline at 37 °C to assess treatment effects on sperm parameters. The suspensions (n = 6) underwent examination for motility, viability, and sperm concentration using established techniques [[33], [34], [35]].
Sperm Count: The sperm count was computed using the WHO laboratory manual [35]. A haemocytometer with enhanced Neubauer ruling was used. Using a WBC pipette, the sperm suspension was diluted 20-fold with normal saline. One drop of the diluted suspension was placed on each side of the haemocytometer and allowed to settle for 30 min in a humid chamber. Spermatozoa were counted in four corner squares using a microscope at 250× magnification.
Spermatozoa Concentration = N X 0.2 × 10^6 [Average number of spermatozoa counted (N) X multiplication factor (10,000) X dilution factor (20)]
Sperm Motility: Sperm motility was investigated immediately after the animals were sacrificed. A drop of sperm suspension was applied on a clean glass slide and covered with a coverslip. The slide was kept at 37 °C for roughly a minute before being inspected with a 400× microscope. Sperm motility was assessed from various perspectives, with any degree of movement termed motile. Motility was calculated as the percentage of motile spermatozoa among the total observed.
Motility (%) = (Number of motile spermatozoa / Total number of spermatozoa) × 100 |
SpermViability: Sperm viability was evaluated using the supra-vital staining technique. Eosine-nigrosine stain was used, and the glassware was kept at 37 °C. A drop of sperm suspension was combined with eosin-nigrosine stain (1% eosinY+ 10% nigrosine; 1:1), and a smear was formed on a clean glass slide. The slide was inspected under a 400× microscope, with viable spermatozoa looking colorless and non-viable ones staining reddish. Viability was calculated as the percentage of viable spermatozoa among the total observed.
Viability (%) = (Number of viable spermatozoa / Total number of spermatozoa) × 100 |
Sperm Morphology: Sperm morphology was examined at 400× magnification by observing sperm suspension smeared onto a fresh glass slide. The criteria of Wyrobek and Bruce (1975) and Zaneveld and Polakoski (1977) [36,37]were employed for evaluation of sperm abnormalities. Approximately, two hundred spermatozoa from each sample were examined for morphological abnormalities, including tail and head abnormalities. Normal sperm morphology was also assessed.
2.6. Daily sperm production (DSP)
To evaluate the effects of Shilajit supplementation on DSP in CPA-exposed mice, elongated spermatids (step 14–16 spermatids) resistant to sonication were counted according to Meistrich and van Beek (1993). In brief, one testis was homogenized in 1ml of chilled distilled water and sonicated for 1.5 min. The sample was counted in a haemocytometer under a phase contrast microscope at 400X magnification. In mice spermatogenesis, developing spermatids spend 4.84 days in step 14–16. Therefore, to calculate daily sperm production, the number of steps 14–16 spermatids was divided by 4.84 [38,39].
2.7. Flow cytometry
To determine the effect of Shilajit supplementation on testicular germ cell dynamics in CPA-exposed mice, we quantified the various germ cell types involved in spermatogenesis using flow cytometric analysis on testicular tissue in accordance with our established methodology [38]. A flow cytometer (Beckman Coulter, CytoFLEX LX, United States) was employed to enumerate cells based on their DNA content, with approximately ten thousand fluorescent events recorded. The resulting DNA histograms were analyzed using CytExpert, Version 2.5 (Beckman Coulter) software. The data obtained from the analysis were presented as percentages of different germ cell populations (1C: round spermatid, 2C: spermatogonia, 4C: primary spermatocytes, S-Ph: S-phase germ cells) and germ cell ratios.
2.8. Histopathology of testis and epididymis
The testes and epididymis were removed and carefully cleaned before being fixed in fresh Bouin's fluid for histological examination in accordance with our established methodology. To assess the effects of CPA exposure and Shilajit treatment on the spermatogenic cycle, we selected 100 cross-sections of seminiferous tubules from five testes of five mice in each group. These were identified and categorised based on the stages of spermatogenesis, as outlined by Russell et al. [40]. In cases of severe impairment where accurate identification was not possible, the seminiferous tubules were assigned to stages I–V, VI–VIII, or IX–XII. Seminiferous tubule diameter and germinal epithelium height in Stage VII seminiferous tubules that were round or slightly oblique (n = 15) were also calculated by Motic software. Additionally, we also calculated the percentage of affected seminiferous tubules [33].
2.9. Immunohistochemical staining of proliferating germ cell nuclear antigen (PCNA)
In this study, immunohistochemistry was used to detect PCNA in testicular sections, following the protocol established by Patel et al. [34]. In brief, 6 μm thick testicular sections were deparaffinized, rehydrated, and rinsed in PBS. Antigen unmasking was achieved by heating the sections in 10mM citrate buffer with a 750W microwave before washing them three times with PBS. To counteract endogenous peroxidase activity, sections were treated with 3% H2O2 in methanol. Subsequently, a 2-h incubation with 5% normal goat serum was performed to prevent nonspecific antibody binding. Sections were then exposed to an anti-PCNA primary antibody (1:300 in PBS with 2% NGS) overnight at 4 °C. Subsequently, sections were treated with an HRP-conjugated goat anti-mouse IgG secondary antibody for 2 h after three PBS washes. Prior to counterstaining with haematoxylin, sections were incubated with DAB substrate (Vector Laboratories, CA, USA). The sections were dehydrated before being mounted with DPX. This experiment also included negative control slides, which contained 2% NGS in PBS instead of the primary antibody. The semi-quantification of PCNA staining in the testes of different treatment groups was performed by the ImageJ software. The DAB stained area for PCNA in the testis was obtained by using threshold tool of ImageJ as described previously [41] and data was presented as the percentage area of PCNA staining.
2.10. Evaluation of oxidative stress
In order to investigate the impact of various treatments on oxidative stress status of testis, a testicular homogenate {10% (w/v)} in 0.01mol/L chilled PBS at pH 7.4 was subjected to centrifugation at 2600rpm at 4 °C for 10 min. To assess lipid peroxidation (LPO), half of the resulting supernatant was employed, following the methodology described by Ohkawa et al. [42].
The remaining half of the supernatant was centrifuged again at 4 °C for 30 min at 12000rpm for 30 min. The supernatant obtained from this step was used to evaluate the activity of superoxide dismutase (SOD), following the protocol outlined by Das et al. [43]. Protein quantification was carried out utilizing Lowry's technique [44].
2.11. Serum hormone assay
Serum testosterone (catalogue number: DKO002) and estradiol (catalogue number: DKO003) levels were determined by ELISA kit (DiaMetra, Italy) following the instructions provided by the manufacturer.
2.12. Immunoblot analysis
To examine the expression of various markers in the testis, including germ cell proliferating markers (PCNA), oxidative stress markers (Keap-1 and Nrf-2), testicular steroidogenesis markers (StAR, CYP11A1, 3β-HSD, 17β-HSD, and CYP-19), apoptotic markers (Bax, Bcl-2, and Caspase-3), and Sertoli cell functional proteins (N-cadherin and β-catenin), immunoblot analysis was performed. Protein quantification and immunoblot analysis were conducted following the established laboratory protocol [45]. The signals were detected using the ChemiDoc system (Analytik Jena). The densitometry of immunoreactive bands was analyzed using AlphaEaseFC software (AlphaImager 2200), with β-actin serving as the loading control. The details of antibodies used in the present study and their dilution are given in Supplementary Table 1.
2.13. Statistical analyses
The data were expressed as the mean ± SEM (Standard Error of the Mean). Statistical analysis was performed using IBM SPSS Statistics 16 for Windows, employing a one-way ANOVA, followed by Duncan's multiple range test for making comparisons between different groups. Statistical significance was defined at p < 0.05.
3. Results
3.1. Body weight and testis weight
Body weights (both initial and final) did not differ significantly between mice exposed to CPA, those treated with Shilajit after CPA exposure, and the control group (p > 0.05, Fig. 1a). In terms of absolute testicular weight, CPA exposure significantly decreased it compared to the control group (*p < 0.05). Conversely, both doses of Shilajit treatment substantially increased the absolute testicular weight in mice exposed to CPA (#p < 0.05, Fig. 1b).
3.2. Sperm analysis and testicular daily sperm production (DSP)
CPA exposure significantly reduces cauda epididymal spermatozoa motility, viability, and count while increasing morphological abnormalities (*p < 0.05). Abnormal tails were the most common sperm morphological abnormalities, followed by abnormal heads (Fig. 1d, Table 1). When compared to CPA-treated mice, both doses of Shilajit treatment resulted in significant improvements in motility, count, and viability, as well as decreased morphological abnormalities of cauda epididymal spermatozoa. (#p < 0.05, Fig. 1c and d and Table 1).
Table 1.
Groups | Sperm morphology |
||
---|---|---|---|
Normal | Head abnormalities | Tail abnormalities | |
Control | 88.6 ± 1.032 | 4.6 ± 0.925 | 6.8 ± 1.06 |
CPA | 42.4 ± 0.929* | 21.4 ± 0.81* | 36.2 ± 0.967* |
CPA + S100 | 55.8 ± 1.245*# | 17.8 ± 0.582*# | 26.4 ± 0.509*# |
CPA + S200 | 58.2 ± 1.16*# | 14.6 ± 0.747*# | 27.2 ± 0.858*# |
Values are mean ± SEM for 6 mice.
* and # indicated significant different from controls (p < 0.05) and CPA-exposed mice (p < 0.05) respectively.
In comparison to the control group, mice exposed to CPA had a notable reduction in testicular daily sperm production (*p < 0.05). Nonetheless, both low and high doses of Shilajit administration led to a substantial increase in testicular daily sperm production among mice exposed to CPA (#p < 0.05, Fig. 1e).
3.3. Flow cytometry of testicular germ cells
The testes of control animals showed three distinct peaks in germ cell populations when the DNA content of various germ cells was examined: spermatogonia (2C), primary spermatocytes (4C), and total round spermatids (1C). The phase between the 4C and 2C peaks, during which cells are actively engaged in DNA synthesis, is commonly referred to as the S-Phase. CPA exposure significantly decreased the number of 1C and 4C populations as well as 1C:2C and 4C:2C germ cell ratios when compared to controls (*p < 0.05), however, it had no effect on the number of 2C and S-Phase cells (p > 0.05, Fig. 2a–c). Shilajit treatment, on the other hand, significantly increased 1C and 4C populations along with an increase in 1C:2C, 1C:4C, 4C:2C and 4C:S-Ph germ cell ratios in CPA-exposed mice (#p < 0.05, Fig. 2a–c).
3.4. Histopathology of testis
The histological examination of the testis in the control mice revealed the presence of a typical and healthy histoarchitecture within the seminiferous tubules (Fig. 3a and 3e-g). However, CPA-exposed mice exhibited significant histopathological changes, including reduced germ cell count, tubule vacuolization, seminiferous epithelium loosening, and germ cell exfoliation into the tubule lumen, mainly in spermatogenic cycle stages I–VIII (Fig. 3b and 3h-j). Treating CPA-exposed mice with low-dose Shilajit improved tubular histoarchitecture by increasing germ cell count (Fig. 3c and 3k-m). Further, when CPA-exposed mice were treated with a high dose of Shilajit, the histoarchitecture of the seminiferous tubules was almost similar to that of the controls (Fig. 3d and 3n-p). Furthermore, quantitative histopathology revealed that both doses of Shilajit treatment significantly increased germinal epithelium height (GEH), seminiferous tubule diameter (STD), and decreased the affected seminiferous tubules compared to CPA-exposed mice (#p < 0.05, Table 2).
Table 2.
Groups | STD (μm) | GEH (μm) | Affected ST (%) |
---|---|---|---|
Control | 298.2 ± 7.69 | 87.70 ± 1.82 | 11.67 ± 1.6 |
CPA | 258.45 ± 5.56* | 68.3 ± 2.02* | 56.60 ± 4.4* |
CPA + S100 | 294.43 ± 4.32# | 85.43 ± 1.71# | 35 ± 2.89*# |
CPA + S200 | 292.23 ± 5.17# | 85.02 ± 1.70# | 31.6 ± 4.4*# |
Values are mean ± SEM for 5 mice.
* and # indicated significant different from controls (p < 0.05) and CPA-exposed mice (p < 0.05) respectively.
3.5. Histopathology of epididymis
In all segments (I–V) of the epididymis, the histological examination of the control mice revealed normal histology (Fig. 4a–e). However, the epididymis of CPA-exposed mice displayed detectable histological changes, including vacuole-like spaces in segments II and IV of the epididymal epithelium and significantly fewer spermatozoa in the lumen of segment V (Fig. 4f–j). Shilajit treatment, in contrast, resulted in normal histoarchitecture in all segments (I–V) and a dose-dependent increase in the number of sperm cells in the lumen of segment V of the epididymis in CPA-exposed mice (Fig. 4k-o; 4p-t).
3.6. Immunohistochemical staining and Western blot analysis of proliferating germ cell nuclear antigen (PCNA)
PCNA immunolocalization in the testis of control mice (Fig. 5a) showed that this protein was expressed in spermatogonia and early spermatocytes. When CPA-exposed mice (Fig. 5b) were compared to controls, there was a significant decrease in PCNA immunostaining (*p < 0.05, Fig. 5f). Low and high doses of Shilajit treatment (Fig. 5c–d respectively), on the other hand, showed a significant increase in PCNA immunostaining in dose-dependent manner in the testis of CPA-exposed mice (#p < 0.05, Fig. 5f). Furthermore, PCNA immunolocalization was also supplemented with Western blot analysis. Western blot analysis of PCNA revealed a significant decrease in PCNA expression in CPA-exposed mice compared to controls (*p < 0.05). By contrast, treatment with both doses of Shilajit significantly increased PCNA expression in CPA-exposed mice (#p < 0.05, Fig. 5g–h).
3.7. Lipid peroxidation and activity of superoxide dismutase
In comparison to controls, mice exposed to CPA showed a significant increase in lipid peroxidation (LPO) levels in testicular tissue (*p < 0.05, Fig. 5i), while superoxide dismutase (SOD) activity was significantly decreased (*p < 0.05, Fig. 5j). Shilajit treatment reduced LPO levels induced by CPA non-significantly (p > 0.05) in the testis while significantly increasing SOD levels compared to CPA-exposed mice (#p < 0.05, Fig. 5i–j).
3.8. Western blot analysis of Keap-1 and Nrf-2
In CPA-exposed mice, the expression of Keap-1 was notably elevated compared to the control group (*p < 0.05). However, when treated with both doses of Shilajit, the expression of Keap-1 significantly decreased in mice exposed to CPA (#p < 0.05). Conversely, Nrf-2 expression was notably decreased in CPA-exposed mice when compared to the control group (*p < 0.05). Nevertheless, treatment with both doses of Shilajit resulted in a significant increase in Nrf-2 expression in mice that had been exposed to CPA (#p < 0.05, Fig. 5j–k).
3.9. Serum testosterone and estradiol level
The serum testosterone levels of the CPA-exposed mice were significantly lower than those of the control group (*p < 0.05). However, treatment with Shilajit demonstrated a dose-dependent effect, leading to an increase in serum testosterone levels in the CPA-exposed mice, and the increase was significant at higher dose of Shilajit (#p < 0.05, Fig. 6a). Serum estradiol levels, on the other hand, did not differ significantly between control and CPA-exposed mice (p > 0.05). However, Shilajit treatment decreased serum estradiol levels in CPA-exposed mice in a dose-dependent manner, and this decrease was significant in animals given a high dose of Shilajit (#p < 0.05, Fig. 6b).
3.10. Western blot analysis of testicular steroidogenic markers
The results of the Western blot analysis indicated consistent StAR expression across all experimental groups. However, in CPA-exposed mice treated with Shilajit, CYP11A1 expression increased while CYP19 expression decreased (#p < 0.05). Furthermore, CPA-exposed mice that were given a high dose of Shilajit had significantly higher levels of 3β-HSD and 17β-HSD expression in their testis than CPA-exposed mice (#p < 0.05, Fig. 6c–d).
3.11. Western blot analysis of apoptotic markers
In CPA-exposed mice compared to controls, Bax and Caspase-3 expression were markedly upregulated, whereas Bcl-2 expression was significantly downregulated (*p < 0.05). In CPA-exposed mice, the administration of Shilajit resulted in the downregulation of Bax and Caspase-3 expression, while concurrently upregulating the expression of Bcl-2 (#p < 0.05). Furthermore, CPA exposure significantly increased the Bax: Bcl-2 ratio (*p < 0.05), whereas, both doses of Shilajit treatment to these mice significantly decreased the Bax: Bcl-2 ratio in a dose-dependent manner (#p < 0.05, Fig. 7a–b).
3.12. Western blot analysis of Sertoli cell functional proteins
Western blot analysis revealed that N-cadherin expression in CPA-exposed mice was significantly lower than in controls (*p < 0.05), whereas β-catenin expression in these mice was comparable to controls (p > 0.05). However, Shilajit treatment increases the expression of both N-cadherin (#p < 0.05) and β-catenin (p > 0.05) in CPA-exposed mice. (Fig. 7c–e).
4. Discussion
Cyclophosphamide (CPA), a commonly used chemotherapeutic drug, induces excessive oxidative stress that damages testicular cells, leading to male infertility that affects the quality of life of cancer survivors [6,30]. Therefore, there is a dire need for a safe and effective natural supplement that protects the testis from its side effects. Thus, the purpose of this study was to see whether Shilajit could protect testis from the side effects of cyclophosphamide, a well-known chemotherapeutic drug.
In Parkes mice, the acute administration of CPA did not result in notable changes in body weight across any of the treatment groups, however, testicular weight was negatively affected in mice exposed to CPA, indicating that CPA has a targeted effect on the male reproductive system [46]. While analyzing reproductive toxicity, the weight of the reproductive organs provides important information about the environment in which the reproductive organs function [47,48]. The increase in testis weight following Shilajit supplementation after CPA exposure suggests that Shilajit supplementation aids in the restoration of the functional environment of the testis, which has been harmed by CPA exposure.
CPA exposure significantly decreased serum testosterone levels in the current study by inhibiting the activities of Δ5 3β-HSD and 17β-HSD enzymes and increasing the activity of CYP19 enzymes. These findings suggest that the decrease in testosterone levels in CPA-exposed mice is caused by the negative impact of CPA on signaling pathways of testosterone biosynthesis via decreasing the secretion of LH from the pituitary gland [[49], [50], [51]]. In contrast, Shilajit supplementation to CPA-exposed mice reverted the repressing effects of CPA on steroidogenesis. This indicated that Shilajit might have stimulatory effects on HPG-axis by regulating the level of pituitary gonadotropins [26,52]. We found that Shilajit stimulates conversion of cholesterol to pregnanolone by stimulating the CYP11A1 enzyme and pregnenolone to testosterone by increasing the activity of Δ5 3β-HSD and 17β-HSD. Aside from this, Shilajit supplementation slows the conversion of testosterone to estradiol by decreasing the activity of the aromatase enzyme. The findings suggest that Shilajit has the ability to restore testicular steroidogenesis and serum testosterone levels in mice exposed to CPA. This is accomplished by increasing vital steroidogenic enzyme activity while decreasing aromatase activity. Furthermore, higher Shilajit dose appear to be particularly effective in mitigating the negative effects of CPA exposure on testosterone production.
In the present study, acute CPA exposure had a negative impact on the histoarchitecture of the testis. The germinal epithelium height and stage VII seminiferous tubules diameter were adversely affected. Intraepithelial vacuolization, exfoliation of germ cells, and loosening of the germinal epithelium were frequently observed in I to VIII stages of seminiferous tubules in CPA-exposed mice. It's worth noting that major spermatogenesis activities take place in stages I-VIII, and Sertoli cells facilitate these key spermatogenic activities by interacting directly with germ cells and controlling the environmental milieu within the seminiferous tubules, and any change in Sertoli cell signaling as well as function affects the structural integrity of the seminiferous tubular epithelium [53,54]. CPA exposure adversely affected the Sertoli cell functioning by affecting the N-cadherin and β-catenin expression. Therefore, the loosening between the germ cell layers and exfoliation of germ cells after CPA exposure in the present experiment might be attributed to Sertoli cell dysfunction associated with compromised germ cell support and maintenance of the blood-testis barrier. The severely affected structure of seminiferous epithelium and functioning of Sertoli cells after CPA exposure as mentioned above, hampers the process of spermiogenesis and spermiation and overall spermatogenesis supported by our flow cytometric findings for the first time [[55], [56], [57]]. CPA exposure severely diminished 1C and 4C populations and 1C:2C, 4C:2C, 4C:S-Ph and 1C:4C ratios in mice showing overall suppression of transformation of spermatogonia to spermatids. Furthermore, the sharp decrease in the 1C population following CPA exposure indicates that post-meiotic cells are highly sensitive to the exposure. These findings are further supported by decreased daily sperm production. PCNA, a well-known biomarker, has been linked to spermatocyte and spermatogonia proliferation [58]. It is possible to explain the decrease in 4C:2C, 4C:S-Ph ratios and testicular daily sperm production after CPA exposure by evaluating the reduced numbers of PCNA-positive germ cells and PCNA expression levels, which indicate that spermatogonia and spermatocyte proliferation have been primarily affected [31]. Shilajit supplementation improves the Sertoli cell functioning and BTB integrity by improving the expression of N-cadherin and β-catenin along with increases in the 1C and 4C population and 1C:2C, 4C:2C, 4C:S-Ph and 1C:4C ratios. Further, increased numbers of PCNA-positive germ cells and PCNA expression levels after Shilajit supplementation at both doses in CPA-exposed mice indicate the stimulation of spermatogonia and spermatocyte proliferation in these animals and it is possible to explain the increase in height of germinal epithelium and testicular daily sperm production after Shilajit supplementation in CPA-exposed animals [52]. Testosterone regulates spermatogenesis by regulating the rate of meiosis and the association of Sertoli cells with germ cells [11]. In the absence of normal testosterone levels, spermatogenesis is slowed and Sertoli cell-germ cell association is disrupted [59]. By inducing steroidogenesis and inhibiting aromatase activity, Shilajit supplementation ensures that enough testosterone is available for normal spermatogenesis in CPA-exposed mice. Shilajit exerts all the above-mentioned effects by acting on local environment of the testis. According to a previous study, its active constituents dibenzo-α pyrones and fulvic acid can penetrate blood-testis barriers reach the target organs such as seminiferous tubules and exert their beneficial effects [52].
The harmony of germ cell proliferation, differentiation, and programmed cell death is essential for mammalian spermatogenesis [60]. The process of programmed cell death known as apoptosis, which takes place to preserve testicular homeostasis, is a characteristic of spermatogenic lineages [60,61]. The Bcl-2 subfamily of programmed cell death modulators promotes cell survival, whereas the Bax subfamily promotes programmed cell death [62]. A significant factor in determining cell fate has been identified as the ratio of Bax to Bcl-2. According to reports, germ cell apoptosis rates are increased by either acute or chronic exposure to CPA via increasing sperm DNA damage [62,63]. In the current study, CPA exposure also caused a disruption in the spermatogenic process by activating the pro-apoptotic factors Bax and caspase 3. The administration of Shilajit, on the other hand, demonstrated a notable reversal of this pattern. This was demonstrated by increased Bcl-2 expression and decreased Bax expression. Therefore, reducing apoptosis by diminishing the Bax/Bcl-2 ratio may also be one of the mechanisms whereby Shilajit improves testicular morphology and functions.
The epididymis is instrumental in the functional maturation of spermatozoa [64]. CPA-induced sperm DNA damage and peroxidation of polyunsaturated fatty acids on spermatozoa membranes had a negative impact on the viability, motility, count, and morphology of cauda epididymal spermatozoa [48]. The decrease in sperm count appears to be the result of the adverse effect of CPA exposure on the testis [47], as we observed mainly exfoliated germ cells and a few spermatozoa in the lumen of segment V of the epididymis [14,65]. Spermatozoa transported from the testes to the epididymis are functionally premature and lack motility [66,67]. Therefore, the decrease in motility and viability of spermatozoa appears to be the result of CPA on the functional environment of the epididymis, as CPA has been reported to adversely affect the functional environment of the epididymis by inducing oxidative stress [30]. CPA is known to increase lipid peroxidation in sperm [14], which is responsible for head and tail abnormalities in sperm, as evident in the current study. The adverse effects of CPA on cauda epididymal sperm motility, viability, count, and morphology were restored by Shilajit supplementation. Shilajit has a positive effect on spermatogenesis, as evidenced by the fact that lumen of segment V is filled with spermatozoa in Shilajit-treated animals, which accounts for the increase in cauda epididymal sperm count. Shilajit improves the functional environment of the epididymis by maintaining the optimum concentration of sialic acid [26,68], which may account for the dose-dependent increase in viability, motility, and a number of morphologically normal spermatozoa by Shilajit supplementation in CPA-exposed animals.
The main consequences of the use of chemotherapeutic drugs have been the disturbed redox balance of tissues by generating excessive oxidative stress in the form of reactive oxygen species (ROS) [62]. Testis is susceptible to damage from oxidative stress, so the antioxidant defence system (Nrf-2/Keap-1 system and superoxide dismutase) offers protection by directly removing free radicals from the testes [69,70]. Under normal circumstances, Nrf-2 is bound to its inhibitor protein, Keap-1, in the cytoplasm. However, during oxidative stress, Keap-1 can become oxidized or undergo covalent modifications, dissociating from Nrf-2. This allows Nrf-2 to regulate lipid peroxidation (LPO) by triggering the expression of various antioxidant enzymes and stimulating the production of superoxide dismutase (SOD), which helps prevent the formation of harmful hydroxyl radicals and enhances cellular antioxidant capacity [71,72]. In the current study, exposure to chemotherapeutic drugs, specifically CPA, resulted in a decrease in Nrf-2 expression and an increase in Keap-1 expression, leading to the suppression of SOD activity. Consequently, this imbalance disrupted the antioxidant defence system and caused oxidative stress, as indicated by an elevated level of lipid peroxidation. Shilajit is a well-known natural antioxidant that has been used in both in vitro and in vivo studies [18,21]. Shilajit at both doses improves the oxidative status of the testis in mice exposed to CPA in the current study. Shilajit increases the expression of Nrf-2, suppresses the expression of Keap-1, and increases the activity of SOD, resulting in a decrease in lipid peroxidation levels. These findings demonstrated that Shilajit had a protective effect against CPA-induced testicular oxidative damage in mice, and that its mechanism may be connected to the activation of the Nrf-2-Keap-1 signaling pathway.
5. Conclusion
In conclusion, CPA exposure exacerbates oxidative imbalance in the testis, resulting in adverse effects on testicular steroidogenesis, Sertoli cell function, germ cell proliferation, and spermiogenesis. This adversely affected overall germ cell dynamics in the testis, as evidenced by an increase in the frequency of affected seminiferous tubules, and adversely affected sperm parameters (motility, viability, count, and morphology) and testicular daily sperm production. In contrast, our study highlights the potent antioxidant properties of Shilajit, which effectively mitigates CPA-induced testicular dysfunction by modulating the Nrf-2/Keap-1 system and restoring oxidative balance, thereby exerting antioxidant effects. Additionally, Shilajit supplementation promotes the expression of crucial steroidogenic enzymes and Sertoli cell functional proteins and facilitates the transformation of spermatogonia into spermatids. It also enhances the expression of PCNA and reduces the Bax/Bcl-2 ratio. Collectively, these effects contribute to the progression of spermatogenesis, leading to notable improvements in testicular daily sperm production and germ cell dynamics. Notably, the 200 mg/kg dose of Shilajit demonstrates significant effectiveness in enhancing steroidogenic markers, testosterone levels, anti-apoptotic protein Bcl-2, and Sertoli cell marker N-cadherin expression. Overall, Shilajit at both doses shows promise in mitigating CPA-induced damage in the male reproductive system, suggesting a potential strategy for managing reproductive dysfunction associated with chemotherapeutic drugs.
Sources of funding
This investigation was supported in part by the ICMR, New Delhi in the form of a major research grant (5/10/FR/29/2019-RBMCH) and by Institute of Eminence research grant (IoE, 6031) from Banaras Hindu University to RKM. AR is recipient of SRF from the CSIR, New Delhi (09/013(0899)/2019-EMR- I).
Author Contributions
Raghav Kumar Mishra: Conceptualization, Methodology, Resources, Writing - Review & Editing, Supervision. Arti Rajpoot: Methodology, Investigation, Writing- Original draft, Data Curation, Visualization, Figures and Tables; Kiran Yadav and Anupam Yadav: Formal analysis, Visualization.
Declaration of Generative AI in Scientific writing
None.
Declaration of competing interest
The authors declare no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Acknowledgement
Authors are thankful to the Indian Council of Medical Research, New Delhi, for the EMR grant (5/10/FR/29/2019-RBMCH) to RKM and the Council of Scientific and Industrial Research, New Delhi, for the JRF (09/013(0899)/2019-EMR-I) to AR. We are also grateful to IoE, Banaras Hindu University, for providing financial support to RKM, and CAS, Department of Zoology, BHU, for extending their lab facilities.
Footnotes
Peer review under responsibility of Transdisciplinary University, Bangalore.
Supplementary data to this article can be found online at https://doi.org/10.1016/j.jaim.2024.100930.
Appendix A. Supplementary data
The following are the Supplementary data to this article:
References
- 1.World Health Organization . 2010. Developing sexual health programmes A framework for action. [Google Scholar]
- 2.Agarwal A., Baskaran S, Parekh N, Cho CL, Henkel R, Vij S., et al. Male infertility. Lancet. 2021;397:319–333. doi: 10.1016/S0140-6736(20)32667-2. [DOI] [PubMed] [Google Scholar]
- 3.Sengupta P., Agarwal A., Pogrebetskaya M., Roychoudhury S., Durairajanayagam D., Henkel R. Role of Withania somnifera (Ashwagandha) in the management of male infertility. Reprod Biomed Online. 2018;36:311–326. doi: 10.1016/J.RBMO.2017.11.007. [DOI] [PubMed] [Google Scholar]
- 4.García-Díaz EC, Gómez-Quiroz LE, Arenas-Ríos E., Aragón-Martínez A., Ibarra-Arias JA, Retana-Márquez MDSI. Oxidative status in testis and epididymal sperm parameters after acute and chronic stress by cold-water immersion in the adult rat. Syst Biol Reprod Med. 2015;61:150–160. doi: 10.3109/19396368.2015.1008071. [DOI] [PubMed] [Google Scholar]
- 5.Liu Q., Lin X., Li H., Yuan J., Peng Y., Dong L., et al. Paeoniflorin ameliorates renal function in cyclophosphamide-induced mice via AMPK suppressed inflammation and apoptosis. Biomed Pharmacother. 2016;84:1899–1905. doi: 10.1016/j.biopha.2016.10.097. [DOI] [PubMed] [Google Scholar]
- 6.Chae YK., Arya A., Iams W., Cruz MR., Chandra S., Choi J., et al. Current landscape and future of dual anti-CTLA4 and PD-1/PD-L1 blockade immunotherapy in cancer; lessons learned from clinical trials with melanoma and non-small cell lung cancer (NSCLC) J Immunother Cancer. 2018;6:1–27. doi: 10.1186/s40425-018-0349-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Salama RM, Abd Elwahab AH, Abd-Elgalil MM, Elmongy NF, Schaalan MF. LCZ696 (sacubitril/valsartan) protects against cyclophosphamide-induced testicular toxicity in rats: role of neprilysin inhibition and lncRNA TUG1 in ameliorating apoptosis. Toxicology. 2020;437 doi: 10.1016/j.tox.2020.152439. [DOI] [PubMed] [Google Scholar]
- 8.Pavin NF., Izaguirry AP, Soares MB, Spiazzi CC, Mendez ASL, Leivas FG., et al. Tribulus terrestris protects against male reproductive damage induced by cyclophosphamide in mice. Oxid Med Cell Longev. 2018;2018 doi: 10.1155/2018/5758191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Li J., You Y., Zhang P., Huang X., Dong L., Yang F., et al. Qiangjing tablets repair of blood-testis barrier dysfunction in rats via regulating oxidative stress and p38 MAPK pathway. BMC Complement Med Ther. 2022;22 doi: 10.1186/s12906-022-03615-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Özbilgin MK, Demirören S., Üçöz M., Oztatlici M. Cyclophosphamide suppresses spermatogenesis in the testis of mice through downregulation of miR-34b and miR-34c. Andrologia. 2021;53 doi: 10.1111/AND.14071. [DOI] [PubMed] [Google Scholar]
- 11.Nayak G., Rao A., Mullick P., Mutalik S., Kalthur SG., Adiga SK., et al. Ethanolic extract of Moringa oleifera leaves alleviate cyclophosphamide-induced testicular toxicity by improving endocrine function and modulating cell specific gene expression in mouse testis. J Ethnopharmacol. 2020;259 doi: 10.1016/j.jep.2020.112922. [DOI] [PubMed] [Google Scholar]
- 12.Ussher JM., Perz J. Infertility-related distress following cancer for women and men: a mixed method study. Psycho Oncol. 2019;28:607–614. doi: 10.1002/PON.4990. [DOI] [PubMed] [Google Scholar]
- 13.Briseño-Bugarín J., Hernández-Ochoa I., Araujo-Padilla X., Mojica-Villegas MA., Montaño-González RI., Gutiérrez-Salmeán G., et al. Phycobiliproteins ameliorate gonadal toxicity in male mice treated with cyclophosphamide. Nutrients. 2021;13 doi: 10.3390/NU13082616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Oyagbemi AA, Omobowale TO, Saba AB, Adedara IA, Olowu ER, Akinrinde AS., et al. Gallic acid protects against cyclophosphamide-induced toxicity in testis and epididymis of rats. Andrologia. 2016;48:393–401. doi: 10.1111/and.12459. [DOI] [PubMed] [Google Scholar]
- 15.Ayza MA, Zewdie KA, Yigzaw EF, Ayele SG, Tesfaye BA, Tafere GG., et al. Potential protective effects of antioxidants against cyclophosphamide-induced nephrotoxicity. Internet J Nephrol. 2022;2022 doi: 10.1155/2022/5096825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Das A., Datta S., Rhea B., Sinha M., Veeraragavan M., Gordillo G., et al. The human skeletal muscle transcriptome in response to oral shilajit supplementation. J Med Food. 2016;19:701–709. doi: 10.1089/jmf.2016.0010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Pandit S., Biswas S., Jana U., De RK., Mukhopadhyay SC., Biswas TK. Clinical evaluation of purified Shilajit on testosterone levels in healthy volunteers. Andrologia. 2016;48:570–575. doi: 10.1111/and.12482. [DOI] [PubMed] [Google Scholar]
- 18.Stohs SJ. Safety and efficacy of shilajit (Mumie, Moomiyo) Phytother Res. 2014;28:475–479. doi: 10.1002/ptr.5018. [DOI] [PubMed] [Google Scholar]
- 19.Bhattacharya SK. Shilajit attenuates streptozotocin induced diabetes mellitus and decrease in pancreatic islet superoxide dismutase activity in rats. Phytother Res. 1995;9:41–44. [Google Scholar]
- 20.Keller JL, Housh TJ, Hill EC, Smith CM, Schmidt RJ, Johnson GO. The effects of Shilajit supplementation on fatigue-induced decreases in muscular strength and serum hydroxyproline levels. J Int Soc Sports Nutr. 2019;16 doi: 10.1186/S12970-019-0270-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Wilson E., Rajamanickam GV, Dubey GP, Klose P, Musial F, Saha FJ., et al. Review on shilajit used in traditional Indian medicine. J Ethnopharmacol. 2011;136:1–9. doi: 10.1016/j.jep.2011.04.033. [DOI] [PubMed] [Google Scholar]
- 22.Cengiz M, Sahinturk V, Yildiz SC., Şahin İK, Bilici N, Yaman SO., et al. Cyclophosphamide induced oxidative stress, lipid per oxidation, apoptosis and histopathological changes in rats: protective role of boron. J Trace Elem Med Biol. 2020;62 doi: 10.1016/j.jtemb.2020.126574. [DOI] [PubMed] [Google Scholar]
- 23.Pant K, Gupta P, Damania P, Yadav AK, Gupta A, Ashraf A., et al. Mineral pitch induces apoptosis and inhibits proliferation via modulating reactive oxygen species in hepatic cancer cells. BMC Compl Alternative Med. 2016;16:1–10. doi: 10.1186/s12906-016-1131-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Jambi EJ., Abdulaziz Alshubaily F. Shilajit potentiates the effect of chemotherapeutic drugs and mitigates metastasis induced liver and kidney damages in osteosarcoma rats. Saudi J Biol Sci. 2022;29 doi: 10.1016/j.sjbs.2022.103393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Mishra RK, Verma HP, Singh N., Singh SK. Male infertility: lifestyle and oriental remedies. Journal of Scientific Research. 2012;56:93–101. [Google Scholar]
- 26.Mishra RK., Jain A., Singh SK. Profertility effects of Shilajit on cadmium-induced infertility in male mice. Andrologia. 2018;50 doi: 10.1111/and.13064. [DOI] [PubMed] [Google Scholar]
- 27.Ayza MA., Balasubramanian R., Berhe AH. Cardioprotective effect of Croton macrostachyus stem bark extract and solvent fractions on cyclophosphamide-induced cardiotoxicity in rats. Evid base Compl Alternative Med. 2020;2020:1–13. doi: 10.1155/2020/8467406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Iqubal A, Syed MA, Najmi AK, Ali J, Haque SE. Ameliorative effect of nerolidol on cyclophosphamide-induced gonadal toxicity in Swiss Albino mice: biochemical-, histological- and immunohistochemical-based evidences. Andrologia. 2020;52 doi: 10.1111/and.13535. [DOI] [PubMed] [Google Scholar]
- 29.Senthilkumar S, Devaki T, Manohar BM, Babu MS. Effect of squalene on cyclophosphamide-induced toxicity. Clin Chim Acta. 2006;364:335–342. doi: 10.1016/j.cca.2005.07.032. [DOI] [PubMed] [Google Scholar]
- 30.Oh MS, Chang MS, Park W., Kim DR, Bae H, Huh Y., et al. Yukmijihwang-tang protects against cyclophosphamide-induced reproductive toxicity. Reprod Toxicol. 2007;24:365–370. doi: 10.1016/j.reprotox.2007.05.007. [DOI] [PubMed] [Google Scholar]
- 31.Kim Y, Lee H, Kim SY, Lim Y. Effects of lespedeza bicolor extract on regulation of AMPK associated hepatic lipid metabolism in type 2 diabetic mice. Antioxidants. 2019;8:599. doi: 10.3390/antiox8120599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Lu WP, Mei XT, Wang Y, Zheng YP, Xue YF, Xu DH. Zn(II)-curcumin protects against oxidative stress, deleterious changes in sperm parameters and histological alterations in a male mouse model of cyclophosphamide-induced reproductive damage. Environ Toxicol Pharmacol. 2015;39:515–524. doi: 10.1016/j.etap.2014.12.014. [DOI] [PubMed] [Google Scholar]
- 33.Mishra RK, Singh SK. Reversible antifertility effect of aqueous rhizome extract of Curcuma longa L. in male laboratory mice. Contraception. 2009;79:479–487. doi: 10.1016/j.contraception.2009.01.001. [DOI] [PubMed] [Google Scholar]
- 34.Patel SK, Singh S, Singh SK. Standardized extract of Bacopa monnieri (CDRI-08): effect on germ cell dynamics and possible mechanisms of its beneficial action on spermatogenesis and sperm quality in male mice. Biochem Biophys Res Commun. 2017;494:34–41. doi: 10.1016/j.bbrc.2017.10.089. [DOI] [PubMed] [Google Scholar]
- 35.World Health Organization . University Press Cambridge; Cambridge: 2010. WHO laboratory manual for the examination and processing of human semen. [Google Scholar]
- 36.Wyrobek A.J., Bruce W.R. Chemical induction of sperm abnormalities in mice. Proceedings of the National Academy of Sciences. 1975;72(11):4425–4429. doi: 10.1073/pnas.72.11.4425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Zaneveld LJD., Polakoski KL. Collection and physical examination of the ejaculate. Techniques of Human Andrology. 1977;1:147–172. [Google Scholar]
- 38.Mishra RK., Singh SK. Safety assessment of Syzygium aromaticum flower bud (clove) extract with respect to testicular function in mice. Food Chem Toxicol. 2008;46:3333–3338. doi: 10.1016/j.fct.2008.08.006. [DOI] [PubMed] [Google Scholar]
- 39.Meistrich ML. Spermatogonial stem cells: assessing their survival and ability to produce differentiated cells. Methods in Reproductive Toxicology. 1993;3:106–123. [Google Scholar]
- 40.Russell L., Ettlin R., Hikim A., Clegg E. Cache River Press; 1990. Histological and histopathological evaluation of the testis. [Google Scholar]
- 41.Jensen EC. Quantitative analysis of histological staining and fluorescence using ImageJ. Anat Rec. 2013;296:378–381. doi: 10.1002/AR.22641. [DOI] [PubMed] [Google Scholar]
- 42.Ohkawa H., Ohishi N., Yagi K. Assay for lipid peroxides in animal tissues by thiobarbituric acid reaction. Anal Biochem. 1979;95:351–358. doi: 10.1016/0003-2697(79)90738-3. [DOI] [PubMed] [Google Scholar]
- 43.Das K, Samanta L, Chainy GBN. A modified spectrophotometric assay of superoxide dismutase using nitrite formation by superoxide radicals. Indian J Biochem Biophys. 2000;37:201–204. [Google Scholar]
- 44.Lowry O., Rosenbrough N., Farr A., Randall R. Protein measurement with the folin phenol reagent. J Biol Chem. 1951;193:265–275. [PubMed] [Google Scholar]
- 45.Yadav K, Yadav A, Rajpoot A, Mishra RK. Postnatal ontogeny of Neuromedin S and its receptors NMUR1 and NMUR2 expression in mouse testis. Peptides. 2023;159 doi: 10.1016/j.peptides.2022.170899. [DOI] [PubMed] [Google Scholar]
- 46.Rezaei S, Hosseinimehr SJ, Zargari M, Karimpour Malekshah A, Mirzaei M, Talebpour Amiri F. Protective effects of sinapic acid against cyclophosphamide-induced testicular toxicity via inhibiting oxidative stress, caspase-3 and NF-kB activity in BALB/c mice. Andrologia. 2021;53 doi: 10.1111/and.14196. [DOI] [PubMed] [Google Scholar]
- 47.Jalali AS, Hasanzadeh S, Malekinejad H. ORIGINAL REPORT Crataegus monogyna aqueous extract ameliorates cyclophosphamide-induced toxicity in rat testis: stereological evidences. Acta Med Iran. 2012;50:1–8. [PubMed] [Google Scholar]
- 48.Torabi F., Malekzadeh Shafaroudi M., Rezaei N. Combined protective effect of zinc oxide nanoparticles and melatonin on cyclophosphamide-induced toxicity in testicular histology and sperm parameters in adult Wistar rats. Int J Reprod BioMed. 2017;15:403–412. [PMC free article] [PubMed] [Google Scholar]
- 49.Ekeleme-Egedigwe CA, Famurewa AC, David EE, Eleazu CO, Egedigwe UO. Antioxidant potential of garlic oil supplementation prevents cyclophosphamide-induced oxidative testicular damage and endocrine depletion in rats. J Nutr Intermed Metab. 2019;18 doi: 10.1016/j.jnim.2020.100109. [DOI] [Google Scholar]
- 50.Abarikwu SO, Otuechere CA, Ekor M, Monwuba K, Osobu D. Rutin ameliorates cyclophosphamide-induced reproductive toxicity in male rats. Toxicol Int. 2012;19:207–214. doi: 10.4103/0971-6580.97224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Potnuri AG., Allakonda L., Lahkar M. Crocin attenuates cyclophosphamide induced testicular toxicity by preserving glutathione redox system. Biomed Pharmacother. 2018;101:174–180. doi: 10.1016/j.biopha.2018.02.068. [DOI] [PubMed] [Google Scholar]
- 52.Biswas TK, Pandit S, Mondal S, Biswas SK, Jana U, Ghosh T., et al. 2010. Clinical evaluation of spermatogenic activity of processed Shilajit in oligospermia; pp. 48–56. [DOI] [PubMed] [Google Scholar]
- 53.Hooley RP, Paterson M, Brown P, Kerr K, Saunders PTK. Intra-testicular injection of adenoviral constructs results in Sertoli cell-specific gene expression and disruption of the seminiferous epithelium. Reproduction. 2009;137:361–370. doi: 10.1530/REP-08-0247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Liu B., ju Shen L., Zhao T xin, Sun M., Wang J ke, lan Long C., et al. Automobile exhaust-derived PM2.5 induces blood-testis barrier damage through ROS-MAPK-Nrf2 pathway in sertoli cells of rats. Ecotoxicol Environ Saf. 2020;189 doi: 10.1016/j.ecoenv.2019.110053. [DOI] [PubMed] [Google Scholar]
- 55.O’Donnell L, Smith LB, Rebourcet D. Sertoli cells as key drivers of testis function. Semin Cell Dev Biol. 2022;121:2–9. doi: 10.1016/j.semcdb.2021.06.016. [DOI] [PubMed] [Google Scholar]
- 56.Liu B., ju Shen L., Zhao T xin, Sun M., Wang J ke, lan Long C., et al. Automobile exhaust-derived PM2.5 induces blood-testis barrier damage through ROS-MAPK-Nrf2 pathway in sertoli cells of rats. Ecotoxicol Environ Saf. 2020;189 doi: 10.1016/j.ecoenv.2019.110053. [DOI] [PubMed] [Google Scholar]
- 57.Yadav A, Yadav K, Rajpoot A, Lal B, Mishra RK. Sub-chronic restraint stress exposure in adult rats: an insight into possible inhibitory mechanism on testicular function in relation to germ cell dynamics. Andrologia. 2022;54 doi: 10.1111/AND.14575. [DOI] [PubMed] [Google Scholar]
- 58.Singh S, Singh SK. Prepubertal exposure to perfluorononanoic acid interferes with spermatogenesis and steroidogenesis in male mice. Ecotoxicol Environ Saf. 2019;170:590–599. doi: 10.1016/j.ecoenv.2018.12.034. [DOI] [PubMed] [Google Scholar]
- 59.Shupe J, Cheng J, Puri P, Kostereva N, Walker WH. Regulation of sertoli-germ cell adhesion and sperm release by fsh and nonclassical testosterone signaling. Mol Endocrinol. 2011;25:238–252. doi: 10.1210/me.2010-0030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Nguyen DH, Soygur B, Peng SP, Malki S., Hu G, Laird DJ. Apoptosis in the fetal testis eliminates developmentally defective germ cell clones. Nat Cell Biol. 2020;22:1423–1435. doi: 10.1038/s41556-020-00603-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Koji T., Shibata Y. Global changes in epigenomes during mouse spermatogenesis: possible relation to germ cell apoptosis. Histochem Cell Biol. 2020;154:123–134. doi: 10.1007/s00418-020-01900-x. [DOI] [PubMed] [Google Scholar]
- 62.Temel Y., Kucukler S., Yıldırım S., Caglayan C., Kandemir FM. Protective effect of chrysin on cyclophosphamide-induced hepatotoxicity and nephrotoxicity via the inhibition of oxidative stress, inflammation, and apoptosis. Naunyn-Schmiedeberg’s Arch Pharmacol. 2020;393:325–337. doi: 10.1007/s00210-019-01741-z. [DOI] [PubMed] [Google Scholar]
- 63.Ghobadi E, Moloudizargari M, Asghari MH, Abdollahi M. The mechanisms of cyclophosphamide-induced testicular toxicity and the protective agents. Expet Opin Drug Metabol Toxicol. 2017;13:525–536. doi: 10.1080/17425255.2017.1277205. [DOI] [PubMed] [Google Scholar]
- 64.Breton S, Ruan YC, Park YJ, Kim B. Regulation of epithelial function, differentiation, and remodeling in the epididymis. Asian J Androl. 2016;18:3–9. doi: 10.4103/1008-682X.165946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Kim W, Kim SH, Park SK, Chang MS. Astragalus membranaceus ameliorates reproductive toxicity induced by cyclophosphamide in male mice. Phytother Res. 2012;26:1418–1421. doi: 10.1002/ptr.4756. [DOI] [PubMed] [Google Scholar]
- 66.Cosentino MJ., Cockett ATK. Review article: structure and function of the epididymis transport of spermatozoa. Urol Res. 1986;14:229–240. doi: 10.1007/BF00256565. [DOI] [PubMed] [Google Scholar]
- 67.Sullivan R., Mieusset R. The human epididymis: its function in sperm maturation. Hum Reprod Update. 2016;22:574–587. doi: 10.1093/humupd/dmw015. [DOI] [PubMed] [Google Scholar]
- 68.Park JS., Kim GY., Han K. The spermatogenic and ovogenic effects of chronically administered Shilajit to rats. J Ethnopharmacol. 2006;107:349–353. doi: 10.1016/j.jep.2006.03.039. [DOI] [PubMed] [Google Scholar]
- 69.Suleiman JB, Nna VU, Zakaria Z, Othman ZA, Eleazu CO, Abu Bakar AB., et al. Protective effects of bee bread on testicular oxidative stress, NF-κB-mediated inflammation, apoptosis and lactate transport decline in obese male rats. Biomed Pharmacother. 2020;131 doi: 10.1016/J.BIOPHA.2020.110781. [DOI] [PubMed] [Google Scholar]
- 70.Yu W., Xu Z., Gao Q., Xu Y., Wang B., Dai Y. Protective role of wogonin against cadmium induced testicular toxicity: involvement of antioxidant, anti-inflammatory and anti-apoptotic pathways. Life Sci. 2020;258 doi: 10.1016/J.LFS.2020.118192. [DOI] [PubMed] [Google Scholar]
- 71.Xue R., Du M., Zhou TY, Ai WZ, Zhang ZS, Xiang XW., et al. Polysaccharides from hemp seed protect against cyclophosphamide-induced intestinal oxidative damage via Nrf2-Keap1 signaling pathway in mice. Oxid Med Cell Longev. 2020;2020 doi: 10.1155/2020/1813798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Fei W., Zhang J., Yu S., Yue N., Ye D., Zhu Y., et al. Antioxidative and energy metabolism-improving effects of Maca polysaccharide on cyclophosphamide-induced hepatotoxicity mice via metabolomic analysis and Keap1-Nrf2 pathway. Nutrients. 2022;14:4264. doi: 10.3390/NU14204264. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.