[go: up one dir, main page]


World J Mens Health. 2024;42:e26. Forthcoming. English.
Published online Feb 14, 2024.
Copyright © 2024 Korean Society for Sexual Medicine and Andrology
Original Article

Effects of Perilla frutescens Var. Acuta in Busulfan-Induced Spermatogenesis Dysfunction Mouse Model

Hyung Jong Nam,1,2,* Min Jung Park,3,4,* Bo Sun Joo,3,4,5 Yean Kyoung Koo,6 SukJin Kim,6 Sang Don Lee,1,7,8 and Hyun Jun Park1,2
    • 1Department of Urology, Pusan National University School of Medicine, Busan, Korea.
    • 2Medical Research Institute of Pusan National University Hospital, Busan, Korea.
    • 3Department of Technical Research, Genoheal, Seoul, Korea.
    • 4Department of R&D Center, The Korea Institute for Public Sperm Bank, Busan, Korea.
    • 5Infertility Institute, Pohang Women’s Hospital, Pohang, Korea.
    • 6Department of R&I Center, COSMAXBIO, Seongnam, Korea.
    • 7Department of Urology, Pusan National University Yangsan Hospital, Yangsan, Korea.
    • 8Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Korea.
Received September 03, 2023; Revised October 30, 2023; Accepted November 05, 2023.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Purpose

The leaves of Perilla frutescens var. acuta (PFA) are generally reported to have antioxidant, anti-allergic, anti-inflammatory, and antitumor effects and commonly used as a traditional medicine in East Asia. This study aimed to investigate the protective effect and antioxidant activity of PFA on busulfan-induced testicular dysfunction, histological damage, oxidative stress (OS), sperm quality, and hormone levels using a mouse model.

Materials and Methods

C57BL/6 male mice were divided into four groups: control, busulfan-only treated, and varying concentrations of PFA (100 and 200 mg/kg) with busulfan. In the busulfan group, 40 mg/kg of busulfan was intraperitoneally injected to induce azoospermia. Mice were orally administered PFA for 35 consecutive days after busulfan administration. Samples were collected and assessed for testis/body weight, testicular histopathology, sperm quality, serum hormone levels, and OS to evaluate the effects of PFA treatment on spermatogenesis dysfunction induced by busulfan.

Results

The busulfan-induced testicular dysfunction model showed reduced testis weight, adverse histological changes, significantly decreased sex hormones and sperm quality, and attenuated OS. These results indicate that PFA treatment significantly increased testis weight, testis/body weight, epididymal sperm count, motility, and testosterone level compared with busulfan alone. PFA treatment also attenuated the busulfan-induced histological changes. Furthermore, compared with mice treated with busulfan alone, PFA supplementation upregulated the testicular mRNA expression of the antioxidant enzymes superoxide dismutase 1 (Sod1) and glutathione peroxidase 1 (Gpx1), with a decrease in malondialdehyde (MDA) production and an increase in SOD and GPx activities.

Conclusions

This study shows that PFA exerts a protective effect against testicular damage by attenuating OS induced by busulfan. Our results suggest that PFA is a potentially relevant drug used to decrease the side effects induced by busulfan on testicular function and sperm during cancer chemotherapy.

Keywords
Antioxidants; Busulfan; Drug therapy; Perilla frutescens; Spermatogenesis

INTRODUCTION

It is known that approximately 15% of all couples have difficulty conceiving and estimated that approximately 20%–70% of these cases are due to male infertility [1, 2]. Overexposure to certain environmental factors and lifestyle behaviors are associated with a decrease in men’s reproductive function and has recently generated some amount of interest [3]. Unfortunately, however, there are more cases of no identifiable reason than cases where a clear cause can be known and treatment can be selected accordingly.

A new term called Male Oxidative Stress Infertility (MOSI) has emerged due to identification of oxidative stress (OS) as an important factor in idiopathic male infertility [4, 5, 6]. In a healthy state, pro-oxidants and antioxidants are balanced, but when the antioxidant defense mechanism is weakened, oxidative damage to gonadal cells and mature spermatozoa is unavoidable [7]. It has been reported that elevated reactive oxygen species (ROS) levels in seminal plasma are found in approximately 30%–40% of patients with male infertility [8]. OS has also been linked to poor fertilization, poor embryonic development, pregnancy loss, birth defects (including autism), and childhood cancer [9, 10]. In a situation where there is no effective treatment for idiopathic male infertility, research to develop antioxidants to lower the OS and use them for treatment has been continuously conducted [11].

Drugs with various antioxidant effects and herbal extracts have been developed in mono- or poly-formulations, and several research results from basic to clinical studies have been published [12, 13]. Antioxidants currently widely used clinically include vitamin A, vitamin C, vitamin E, carnitine, N-acetyl cysteine, coenzyme Q10, and lycopene, along with important antioxidant cofactors, such as zinc, selenium, and folic acid [14]. However, since the effects of these antioxidants are not consistent between reported studies, it is necessary to continuously try to develop new antioxidants [11, 15]. Antioxidants extracted from herbs are relatively safe and have high drug compliance; therefore, they can be high-value candidates for development.

The leaves of Perilla frutescens var. acuta (PFA) are an herbal medicine that has been used for various medical purposes in Asia [16, 17]. It is also widely used as a food ingredient [18]. Studies on PFA have revealed that PFA has antioxidant, anti-glycosuria, anti-inflammatory, anti-allergic, antimicrobial, anti-dementia, antidepressive, antitumor, and anti-obesity properties [16, 17, 18, 19]. The known chemical compounds of this plant include phenolic compounds, flavonoids, anthocyanins, essential oils, and phenylpropanoids [17, 18]. Despite the antioxidant effects of PFA, there have been no reports on the effect of PFA on male fertility. Therefore, we conducted this study to evaluate the therapeutic effects of PFA on spermatogenesis in busulfan-induced spermatogenesis dysfunction in mice.

MATERIALS AND METHODS

1. Reagent and chemicals

The PFA aqueous extract used in this study was supplied by COSMAX Bio. Busulfan (1,4-butanediol dimethanesulfonate) was purchased from Sigma-Aldrich and dissolved in dimethyl sulfoxide (Sigma-Aldrich). Fresh working solutions of busulfan are prepared daily for animal administration owing to poor solubility and prevention of precipitation. Immediately before injection, an equal volume of sterile water was added at room temperature. The busulfan dose was assigned on the basis of previous studies that demonstrated the toxic effect of busulfan on the testes [20] and dosage of PFA was selected based on previous reports demonstrating its anti-oxidative effect [17].

2. Animals and experimental design

10-week-old C57BL/6J male mice (25.7±1.1 g) were purchased from the Koatech Inc. The animals were allowed free access to sterilized food and water. The mice were housed under standard laboratory conditions and acclimatized for 1 week before the start of experiment. To induce azoospermia in mice, considering the average elimination half-life of busulfan of 2–3 hours, 40 mg/kg of busulfan was injected intraperitoneally into mice twice within 1 day and re-administered after 21 days.

The study mice were randomly divided into as follows: (1) control group (n=10); mice were administered the vehicle only as no injection of busulfan, (2) busulfan group (n=10); mice were injected a dose of 40 mg/kg busulfan intraperitoneally, (3) busulfan+PFA 100 group (n=10); mice were orally administered 100 mg/kg of PFA daily for 35 consecutive days after busulfan injection, (4) busulfan+PFA 200 group (n=10); mice were orally administered 200 mg/kg of PFA daily for 35 consecutive days after busulfan injection. All animal body and testis weights were recorded at the end of the experiment.

3. Organ removal and tissue processing

At the end of the treatment period, all mice were euthanized using carbon dioxide asphyxiation, and the testes were excised quickly from each mouse. Testicular tissues were divided into two samples. One testis was fixed in Bouin’s solution for paraffin blocks for histological examination. The other testis was frozen at -80 ℃ for subsequent biochemical analysis experiments, including assay kits and quantitative polymerase chain reaction (qPCR). Epididymal sperm was also collected from cauda epididymides.

4. Histological examination of the testis

The fixed testicle samples were dehydrated in an ascending series of alcohol, cleared in xylene, embedded in paraffin, and sectioned at 5 µm. The sections were stained with hematoxylin and eosin, and were observed under light microscope (Nikon E100; Nikon) for histopathological evaluation. The diameters of seminiferous tubules (STs) and the cellular epithelium (CE) were evaluated with a calibrated linear scale using ImageScope software (Aperio Technologies). Twenty sections of circular or nearly circular tubules were randomly chosen and examined. The diameters of the seminiferous tubule and germinal epithelium thicknesses were measured across minor and major axes and averaged.

5. Semen analysis

To evaluate sperm parameters, sperm were collected from freshly excised epididymis and incubated in phosphate-buffered saline (Sigma-Aldrich) at 37 ℃ for 10 minutes to allow spermatozoa to swim out. The epididymal sperm count was placed on a Neubauer Chamber (HBG Company) under a coverslip and observed under a light microscope at ×200. The motile sperm percentages and sperm counts (expressed as ×106/mL) were determined under the microscope. Sperm density was counted three times per sample, and the results were averaged. Sperm motility is expressed as the percentage of sperm that showed a progressive movement.

6. Measurement of testosterone and follicle-stimulating hormone

Blood samples obtained from the heart were centrifuged at 3,000 rpm for 20 minutes. The serum testosterone and follicle-stimulating hormone (FSH) levels were measured using enzyme-linked immunosorbent assay kits (Cusabio Biotech Co.) according to the manufacture’s protocols. Each test was performed in triplicate, and three independent experiment results were used for statistical analysis.

7. Evaluation of oxidative stress

Frozen testes were homogenized in ice-cold physiological saline solution with an automatic homogenizer. The levels of malondialdehyde (MDA) and enzymatic activities of superoxide dismutase (SOD) and glutathione peroxidase (GPx) were measured using commercially available kits (Cusabio Biotech Co.) according to the manufacturer’s protocols. Each test was performed in triplicate, and two independent experiments were used for statistical analysis.

8. Quantitative real-time polymerase chain reaction assay

Total RNA was extracted from the frozen testicular tissue using TRIzol reagent (Invitrogen). First-strand cDNA was synthesized using the GoScript Reverse Transcription System (Promega Corporation). Quantitative real-time PCR was performed on an ABI Prism 7500 detection system (PE Applied Biosystems) using the SYBR Green PCR Master Mix (TaKaRa). The primers used for gene amplification are listed in Table 1. The expression ratios were calculated using the comparative CT method (2-ΔΔCt). The expression of target genes was determined relative to β-actin as an internal control to determine the relative expression of target genes.

Table 1
Primers sequences used for real time polymerase chain reaction amplification

9. Statistical analysis

All results are expressed as mean±standard deviation. Data were analyzed using Excel’s paired t-test to compare the means of the two samples. Statistical significance was accepted for p-values <0.05.

10. Ethics statement

All animal experiments performed in the present study were approved by the Institutional Animal Care and Use Committee (IACUC) of Pusan National University Hospital (IACUC approval No. PNUH-2021-184) and complied with Guidelines for Care and Use of Laboratory Animals.

RESULTS

1. Effect of Perilla frutescens var. acuta on body and testis weights in busulfan-treated mice

Body weight and absolute and relative testicular weights were significantly lower in the busulfan group than in the control group (Fig. 1 and Table 2) (p<0.01). In contrast, the body weight and absolute and relative testicular weights of the busulfan+PFA 200 group were significantly higher than those of the busulfan group (p<0.05).

Fig. 1
Effect of Perilla frutescens var. acuta (PFA) on body and testis weight in busulfan-treated mice. (A) Body weight. (B) Absolute testis weight. (C) Testis/body weight. Treatment with busulfan alone was significantly affected the body and testis weight of mice, whereas PFA administration with busulfan partially restored the body and testis weight. All value are expressed as mean±standard deviation (n=10). ap<0.01 vs. control group; bp<0.05 vs. busulfan group.

Table 2
Reproductive organ weights, seminiferous tubular diameter in all groups

2. Effect of Perilla frutescens var. acuta on busulfan-induced changes in testicular histology

Busulfan treatment resulted in obvious morphological alterations in the testes of mice, including atrophy of the STs, disorganization of the CE, absence of spermatozoa, depletion of spermatogonial cells, and detachment of germ cells (Fig. 2A). However, all of these adverse histological changes induced by busulfan were significantly attenuated by the oral administration of PFA. Typical seminiferous tubular structures with normal arrangement of spermatogenic cells were observed in mice orally administered PFA 100 and 200 mg/kg as well as control mice.

Fig. 2
Effect of Perilla frutescens var. acuta (PFA) on busulfan-induced changes in testicular histology. (A) Hematoxylin and eosin staining of the seminiferous tubules in the four groups (magnification 200 µm). Photomicrograph of seminiferous tubules in the control group had a typical fully normal appearing spermatogenesis activity in seminiferous tubules. In the busulfan group, it had marked atrophy of the seminiferous tubules with germinal epitheliums aplasia. Spermatid (arrow), diameter of cellular epithelium (CE) (black bar), and Sertoli cells (asterisks). (B, C) The diameter of the seminiferous tubules and germinal epithelium height. All values are expressed as mean±standard deviation (n=10). ap<0.001 vs. control group; bp<0.001 vs. busulfan group.

The diameters of total STs and CE per testis in the busulfan group decreased compared with those in the control group (p<0.001). However, the diameters of ST and CE were significantly higher in the PFA-treated group than in the busulfan group (p<0.001). No significant differences in the diameter of ST and CE were found between the 100 and 200 mg/kg of PFA groups (Fig. 2B, 2C and Table 2).

3. Effect of Perilla frutescens var. acuta on epididymal sperm count and motility in busulfan-treated mice

Epididymal sperm count and sperm motility percentages were significantly lower in the busulfan group than in the control group (17.5±3.1×106/mL vs. 36.0±5.5×106/mL and 23.9%±5.0% vs. 48.8%±4.3%, respectively) (p<0.001). Compared with busulfan group, the oral administration of 100 and 200 mg/kg of PFA in mice treated with busulfan resulted in a significant increase in epididymal sperm count (26.0±2.6×106/mL and 33.3±2.4×106/mL, respectively) (p<0.001) (Fig. 3A). However, only the 200 mg/kg PFA group had a markedly increased epididymal sperm motility compared with the busulfan group (37.0%±3.5% and 23.9%±5.0%, respectively) (Fig. 3B).

Fig. 3
Effect of Perilla frutescens var. acuta (PFA) on epididymal sperm count and motility in busulfan-treated mice. (A) Total sperm counts. (B) Sperm motilities. Treatment of mice with busulfan alone caused a decrease in sperm number and motility, whereas PFA administration with busulfan significantly restored sperm number and motility. Only 200 mg/kg of PFA administration significantly increased sperm motility. All values are expressed as mean±standard deviation (n=10). ap<0.001 vs. control group; bp<0.01 vs. busulfan group.

4. Effect of Perilla frutescens var. acuta on busulfan-induced defective serum hormone levels

The serum testosterone and FSH levels were both significantly decreased in the busulfan group compared with those in the control group (Fig. 4) (p<0.001). However, PFA treatment significantly ameliorated the busulfan-induced abnormal serum testosterone hormone levels (p<0.01), whereas no significant difference was observed in the serum FSH levels between the busulfan and busulfan+PFA groups.

Fig. 4
Effect of Perilla frutescens var. acuta (PFA) on busulfan-induced defective serum hormone levels. (A) Serum testosterone levels, (B) serum follicle-stimulating hormone (FSH) levels. Treatment of mice with busulfan alone caused a decrease of serum testosterone and FSH concentration, whereas PFA administration with busulfan significantly restored serum testosterone concentration. All values are expressed as mean±standard deviation (n=10). ap<0.001 vs. control group; bp<0.01 vs. busulfan group.

5. Effect of Perilla frutescens var. acuta on busulfan-induced testicular oxidative stress

Biochemical analysis of testicular SOD, GSH, and MDA was performed to evaluate the anti-oxidative effect of PFA on busulfan-induced testicular OS. After exposure to busulfan, the activities of SOD and GPx were significantly decreased in the testis relative to the control group, and the formation of MDA was significantly increased (p<0.001). However, PFA administration significantly reduced the production of MDA and enhanced the activities of SOD and GPx in the testes of mice exposed to busulfan (p<0.01) (Fig. 5A–5C). Compared with the busulfan group, PFA treatment significantly increased the activity of SOD and decreased the MDA level in a dose-dependent manner in the testes of mice (Fig. 5A, 5C).

Fig. 5
Effect of Perilla frutescens var. acuta (PFA) on oxidative stress in the testes of busulfan-treated mice. Testicular antioxidant enzymatic activities ([A] SOD and [B] GPx) and MDA (C) contents. mRNA expression of Sod1 (D) and Gpx1 (E). Testicular SOD, and GPx activities significantly decreased following busulfan treatment as compared with the control group; however, PFA administration restored these activities. By contrast, testicular MDA levels significantly increased following busulfan treatment and significantly decreased after PFA administration. Busulfan treatment significantly downregulated Sod1, and Gpx1 expression, all of which were subsequently significantly upregulated following PFA treatment. Each polymerase chain reaction was performed in duplicate on each sample. Relative gene expression levels were calculated vs. β-actin (n=10). All values are expressed as mean±standard deviation. SOD: superoxide dismutase, GPx: glutathione peroxidase, MDA: malondialdehyde. ap<0.001 vs. control group; bp<0.01 vs. busulfan group.

qPCR analysis revealed a significant downregulation of genes encoding antioxidant enzymes Sod1 and Gpx1 in busulfan-treated animals relative to controls (p<0.001) (Fig. 5D, 5E). However, PFA administration caused a significant upregulation (p<0.01) in Sod1 and Gpx1 expression (Fig. 5D, 5E).

DISCUSSION

In the present study, busulfan-induced testicular injury in mice was characterized by decreased testis/body weight, decreased epididymal sperm count and motility, and disorganized seminiferous epithelium, which may be a consequence of elevated OS in the testis and abnormal hormone levels. To the best of our knowledge, this is the first study to demonstrate the beneficial effects of PFA in busulfan-related testicular injury by attenuating OS in the male reproductive system. The results of this study showed that sperm concentration and motility were significantly increased in male mice with busulfan-induced testicular toxicity when administered with 200 mg/kg of PFA compared with those with busulfan alone. The level and expression of SOD and GPx, the diameter of the germinal epithelium, and testosterone hormone levels were increased in busulfan-induced male infertile mice administered 100 and 200 mg/kg of PFA compared with the busulfan alone group.

The incidence of cancer continues to increase and one of the most prevalent cancers during reproductive age is leukemia [21, 22]. Despite the development of cancer treatments such as chemotherapy regimens, radiotherapy therapy, and surgical options, busulfan is known for its effective management of leukemia, especially in children. However, because busulfan is very effective in spermatogonial stem cells (SSCs), it affects the male reproductive system and causes loss of fertility, especially in young cancer survivors [20]. In addition, busulfan-induced male sterility in mice is very similar to that in humans [20].

Among various approaches as the animal model in infertility studies, 5 to 40 mg/kg of busulfan was the most commonly used for induction of infertility mice due to its easy administration by intraperitoneal injection [23]. Zohni et al [24] reported that single injections of busulfan at 15, 30, or 45 mg/kg were direct consequences of SSC loss. Wang et al [23] reported that a busulfan dose of up to 30 mg/kg was optimal in terms of reducing lethality in BALB/c mice. Dobrinski et al [25] reported that the intraperitoneal injection of busulfan at 50 mg/kg was lethal in BALB/c mice due to hematopoietic toxicity. Xie et al [20] reported that testes showed dynamic changes at 9 days after the administration of 40 mg/kg of busulfan but partially recovered 90 days after busulfan administration. To date, there has not yet been a systematic study to determine the optimal dose of busulfan for the induction of infertility in experimental animals such as mice. In previous studies, the intraperitoneal injection of busulfan at 40 mg/kg in C57BL/6 mice resulted in decreased testis/body weight, atrophied STs, reduced sperm function, and abnormal hormone levels than 10 to 20 mg/kg. Consistent with previous studies, the results of this study also showed lower sperm quantity and quality in mice treated with higher busulfan doses than in those treated with lower busulfan doses. However, the mortality rate of a single intraperitoneal injection of 40 mg/kg of busulfan was up to 15% for 1 week due to the possibility of the inhibition of hematopoiesis by absorption into other organs via blood (data not shown). In the current investigation, we selected two intraperitoneal injections of busulfan at a total dose of 40 mg/kg at 3-hour interval to establish a model of azoospermia with low hematopoietic toxicity and zero mortality. In the present study, with no mortality, significantly lower body and testicular weights were observed in the busulfan-treated group than in the control group (Fig. 1).

Several pharmacological studies have examined the leaves of PFA for their antioxidant, anti-inflammatory, anti-allergic, antimicrobial, and antitumor effects [17, 18]. Kim et al [17] reported the efficacy of the antioxidant activity of PFA in polyphenolic components such as rosmarinic acid, luteolin-7-O-glucuronide, and apigenin-7-O-glucuronide. With reference to previous studies on the antioxidant function of PFA, our experimental study demonstrated the relationship between the antioxidant effects of PFA and busulfan-induced testicular damage due to OS, which is very important in sperm fertility potential. Building on previous studies on the antioxidant functions of PFA, our experimental study investigated the antioxidant effects of PFA and the restoration of sperm quality in mice with busulfan-induced testicular injury as an ex vivo effect of antioxidant defense. The present study demonstrated, for the first time, the therapeutic effects of PFA on busulfan-induced testicular damage by suppressing OS. According to the histomorphometric findings, the control group showed complete spermatogenic activity, and in the busulfan group, the thickness of the germinal epithelium decreased due to the decrease in the proportion of spermatogonia, spermatocytes, and spermatids (Fig. 2). PFA administration in busulfan-treated mice significantly increased the proportion of spermatogonia and spermatocytes compared with busulfan alone. Based on our data, we suggest that PFA administration protects germ cells from busulfan-induced testicular toxicity. Zhengwei et al [26], reported that a direct relationship between the germinal cell number and testis weight in primates. Our findings are consistent with previous reports showing that busulfan-induced testicular weight loss is associated with loss of germ cells and that testicular weight gain with PFA administration is associated with germ cell recovery (Fig. 1). Quantitative recovery of germ cells can be evidenced by an increase in the total diameter and cellular diameter of the ST, which comprise a substantial portion of the testes. In addition, our results clearly showed that PFA administration increased sperm concentration and motility compared with busulfan alone, which suggests that PFA rescues spermatogenesis (Fig. 3).

OS has also emerged as a major cause of male fertility in at least 40% of the patients with potential impacts on the developmental capacity of embryos and the health and well-being of the offspring [27, 28]. Busulfan is known to partially eliminate stem cells due to its alkylating properties and inhibit the spermatogenesis process, especially by oxidative damage [23, 24]. Testicular OS is known to increase the MDA levels in testicular tissue, while decreasing the activity of protective antioxidant enzymes (SOD and GPx). In this study, busulfan exposure induced increased MDA production along with a decreased activity of the antioxidant enzymes SOD and GPx in the testes of mice, presumably due to testicular damage caused by overproduction of ROS and lack of antioxidant defenses. However, the increased formation of MDA and decreased activities of SOD and GPx were significantly restored by simultaneous treatment with PFA, suggesting that PFA has an ameliorating effect on busulfan-induced reproductive and developmental toxicity in mice. Our results also showed that PFA administration upregulated the expression of genes encoding the antioxidant enzymes Sod1 and Gpx1 (Fig. 5). However, the mechanisms involved in PFA-mediated OS suppression are not yet clear; therefore, further mechanistic studies of the relationship between PFA and transcription factors affecting antioxidants are needed.

In this study, the lowered serum testosterone and FSH levels in mice treated with busulfan injection in male mice were consistent with previous reports [29]. It is now recognized that Leydig cells produce testosterone in response to LH, which diffuses into the STs and acts as a paracrine factor necessary to maintain spermatogenesis [30]. Although the mechanism underlying the detrimental effects of busulfan injection is not yet clear, previous studies have shown that injection of high-dose busulfan inhibits testosterone production by affecting the exocrine and endocrine compartments of the testes with decreased antioxidant enzyme activity, increased testicular lipid peroxidation, and persistent impairment of Leydig cell function [30]. The oral administration of PFA in busulfan-treated mice significantly increased the serum testosterone levels compared with the busulfan-treated group (Fig. 4). Therefore, the amelioration in the serum testosterone levels observed after PFA administration in busulfan-treated mice may be related to the recovery of Leydig cells by ROS inhibition.

In the present study, the body and testicular weights, sperm count, sperm motility, serum testosterone levels, increased SOD levels, and decreased MDA levels occurred in a dose-dependent manner in the PFA- and busulfan-treated groups compared with those in the busulfan alone group. These effects of the high dose of PFA were better than those of low doses during the administration period.

The present study has some limitations. First, there are no existing standards for optimal oral antioxidant supplementation regimens and duration of administration; therefore, further investigation of various dosages or long-term administration of PFA is needed. Second, it is necessary to evaluate the ability of PFA to inhibit apoptosis in busulfan-induced testicular tissue because spermatogenesis is a complex process and germ cell apoptosis is essential. Third, we could not investigate the effects of each of the individual components of PFA. An in-depth study of the active ingredients of PFA should follow.

The oral administration of PFA restored busulfan-induced disrupted spermatogenesis by improving testicular germ cell development and sperm quality through the attenuation of OS. These benefits of PFA can be used to improve male reproduction in patients treated with busulfan or other cancer medications or in patients with idiopathic failed gametogenesis. We propose that PFA may affect these infertile couples through improved spermatogenesis.

CONCLUSIONS

In conclusion, PFA exhibited a protective effect against busulfan-induced testicular damage in male mice, showing decreased sperm count and motility, testosterone levels, and antioxidant status and increased MDA levels. Our results also showed that the aforementioned changes would be dose-dependent in PFA. Our results suggest that PFA helps improve the reproductive parameters in busulfan-induced male reproductive toxicity.

Notes

Conflict of Interest:The authors have nothing to disclose.

Funding:None.

Author Contribution:

  • Conceptualization: HJP, SDL.

  • Data curation: HJN, MJP.

  • Formal analysis: HJN, MJP, BSJ.

  • Funding acquisition: HJN.

  • Investigation: HJN, MJP, BSJ.

  • Methodology: BSJ, YKK, SJK.

  • Project administration: MJP.

  • Resources: YKK, SJK.

  • Software: YKK, SJK.

  • Supervision: HJP, SDL.

  • Validation: BSJ, HJP.

  • Visualization: HJN, MJP, BSJ.

  • Writing – original draft: HJN, MJP.

  • Writing – review & editing: HJP.

Data Sharing Statement

The data that support the findings of this study are available from the corresponding author upon reasonable request.

References

    1. Trussell JC. Optimal diagnosis and medical treatment of male infertility. Semin Reprod Med 2013;31:235–236.
    1. Agarwal A, Mulgund A, Hamada A, Chyatte MR. A unique view on male infertility around the globe. Reprod Biol Endocrinol 2015;13:37
    1. Leisegang K, Dutta S. Do lifestyle practices impede male fertility? Andrologia 2021;53:e13595
    1. Agarwal A, Parekh N, Panner Selvam MK, Henkel R, Shah R, Homa ST, et al. Male oxidative stress infertility (MOSI): proposed terminology and clinical practice guidelines for management of idiopathic male infertility. World J Mens Health 2019;37:296–312.
    1. Hamada A, Esteves SC, Nizza M, Agarwal A. Unexplained male infertility: diagnosis and management. Int Braz J Urol 2012;38:576–594.
    1. Agarwal A, Virk G, Ong C, du Plessis SS. Effect of oxidative stress on male reproduction. World J Mens Health 2014;32:1–17.
    1. Henkel RR. Leukocytes and oxidative stress: dilemma for sperm function and male fertility. Asian J Androl 2011;13:43–52.
    1. Lanzafame FM, La Vignera S, Vicari E, Calogero AE. Oxidative stress and medical antioxidant treatment in male infertility. Reprod Biomed Online 2009;19:638–659.
    1. Buhling K, Schumacher A, Eulenburg CZ, Laakmann E. Influence of oral vitamin and mineral supplementation on male infertility: a meta-analysis and systematic review. Reprod Biomed Online 2019;39:269–279.
    1. Tremellen K. Oxidative stress and male infertility--a clinical perspective. Hum Reprod Update 2008;14:243–258.
    1. Agarwal A, Leisegang K, Majzoub A, Henkel R, Finelli R, Panner Selvam MK, et al. Utility of antioxidants in the treatment of male infertility: clinical guidelines based on a systematic review and analysis of evidence. World J Mens Health 2021;39:233–290.
    1. Majzoub A, Agarwal A. Systematic review of antioxidant types and doses in male infertility: benefits on semen parameters, advanced sperm function, assisted reproduction and live-birth rate. Arab J Urol 2018;16:113–124.
    1. Smits RM, Mackenzie-Proctor R, Yazdani A, Stankiewicz MT, Jordan V, Showell MG. Antioxidants for male subfertility. Cochrane Database Syst Rev 2019;3:CD007411
    1. Adewoyin M, Ibrahim M, Roszaman R, Isa MLM, Alewi NAM, Rafa AAA, et al. Male infertility: the effect of natural antioxidants and phytocompounds on seminal oxidative stress. Diseases 2017;5:9
    1. Keskes-Ammar L, Feki-Chakroun N, Rebai T, Sahnoun Z, Ghozzi H, Hammami S, et al. Sperm oxidative stress and the effect of an oral vitamin E and selenium supplement on semen quality in infertile men. Arch Androl 2003;49:83–94.
    1. Lee CW, Choi HM, Kim SY, Lee JR, Kim HJ, Jo C, et al. Influence of Perilla frutescens var. acuta water extract on the shelf life and physicochemical qualities of cooked beef patties. Korean J Food Sci Anim Resour 2015;35:389–397.
    1. Kim J, Kang H, Choi H, Jo A, Oh DR, Kim Y, et al. Aqueous extract of Perilla frutescens var. acuta relaxes the ciliary smooth muscle by increasing NO/cGMP content in vitro and in vivo. Molecules 2018;23:1777
    1. Jun HI, Kim BT, Song GS, Kim YS. Structural characterization of phenolic antioxidants from purple perilla (Perilla frutescens var. acuta) leaves. Food Chem 2014;148:367–372.
    1. Banno N, Akihisa T, Tokuda H, Yasukawa K, Higashihara H, Ukiya M, et al. Triterpene acids from the leaves of Perilla frutescens and their anti-inflammatory and antitumor-promoting effects. Biosci Biotechnol Biochem 2004;68:85–90.
    1. Xie Y, Deng CC, Ouyang B, Lv LY, Yao JH, Zhang C, et al. Establishing a nonlethal and efficient mouse model of male gonadotoxicity by intraperitoneal busulfan injection. Asian J Androl 2020;22:184–191.
    1. You W, Henneberg M. Cancer incidence increasing globally: the role of relaxed natural selection. Evol Appl 2017;11:140–152.
    1. Michailov Y, Lunenfeld E, Kapelushnik J, Huleihel M. Leukemia and male infertility: past, present, and future. Leuk Lymphoma 2019;60:1126–1135.
    1. Wang DZ, Zhou XH, Yuan YL, Zheng XM. Optimal dose of busulfan for depleting testicular germ cells of recipient mice before spermatogonial transplantation. Asian J Androl 2010;12:263–270.
    1. Zohni K, Zhang X, Tan SL, Chan P, Nagano MC. The efficiency of male fertility restoration is dependent on the recovery kinetics of spermatogonial stem cells after cytotoxic treatment with busulfan in mice. Hum Reprod 2012;27:44–53.
    1. Dobrinski I, Avarbock MR, Brinster RL. Transplantation of germ cells from rabbits and dogs into mouse testes. Biol Reprod 1999;61:1331–1339.
    1. Zhengwei Y, McLachlan RI, Bremner WJ, Wreford NG. Quantitative (stereological) study of the normal spermatogenesis in the adult monkey (Macaca fascicularis). J Androl 1997;18:681–687.
    1. Aitken RJ, Baker MA. The role of genetics and oxidative stress in the etiology of male infertility-a unifying hypothesis? Front Endocrinol (Lausanne) 2020;11:581838
    1. Aitken RJ. Impact of oxidative stress on male and female germ cells: implications for fertility. Reproduction 2020;159:R189–R201.
    1. Mohammadghasemi F, Soleimanirad J, Ghanbari AA. An ultrastructural study on the apoptotic features of spermatogenic cells following busulfan treatment in adult mice. J Reprod Infertil 2008;8:319–329.
    1. Soleimanzadeh A, Mohammadnejad L, Ahmadi A. Ameliorative effect of Allium sativum extract on busulfan-induced oxidative stress in mice sperm. Vet Res Forum 2018;9:265–271.

Metrics
Share
Figures

1 / 5

Tables

1 / 2

PERMALINK