Article
Melatonin provides protection against cisplatin-induced ovarian damage and loss of fertility in mice

https://doi.org/10.1016/j.rbmo.2020.10.001 Get rights and content

Abstract

Research question

Can melatonin provide non-invasive ovarian protection against damage caused by cis-diamminedichloroplatinum (cisplatin) and preserve fertility in female cancer patients? And if so, what is the possible mechanism?

Design

Athymic BALB/c nude tumour-bearing female mice were used to demonstrate whether melatonin affects the antineoplastic effect when co-administrated with cisplatin. Sexually mature and newborn C57BL/6 female mice were used to evaluate the potential effects of melatonin on the ovarian follicle pool, pregnancy rate and litter number in cisplatin-treated mice. The ovaries underwent immunohistochemical, TdT (terminal deoxynucleotidyl transferase)-mediated dUTP nick-end labelling (TUNEL) and gene array analysis to explore the underlying mechanism. In addition, granulosa cells were isolated to investigate the potential protective mechanism of melatonin.

Results

Melatonin not only enhanced the anti-cancer effect of cisplatin in tumour-bearing nude mice, but also reduced ovarian toxicity and preserved long-term fertility in cisplatin-treated C57BL/6 female mice. When co-administrated, melatonin was able to reduce the DNA damage and toxic effects on lipid peroxidation in the ovaries caused by cisplatin. Specifically, melatonin was able to largely restore lipid peroxidation in granulosa cells and thus prevent ovarian follicles from being depleted.

Conclusions

Melatonin has the potential to be used as a chemotherapeutic adjuvant to simultaneously improve the outcome of anti-cancer treatment and preserve ovarian function during cisplatin chemotherapy. Notably, its properties of DNA protection and antioxidant effects on follicles may benefit female cancer survivors and prevent premature ovarian failure as well as fertility loss caused by chemotherapy.

Introduction

Improved screening procedures for the earlier detection and diagnosis of cancers as well as improved anti-cancer therapies have improved the prognosis and prolonged the survival of cancer patients (Noone et al., 2017). However, anti-cancer therapy is often detrimental to fertility and reproductive endocrine function. For female cancer patients, especially young patients with a strong desire for childbearing who need to be treated with gonadotoxic chemotherapeutic agents, new problems such as loss of fertility and iatrogenic premature ovarian failure must be considered (Blumenfeld, 2003).

Much effort has been applied to solving the reproductive problems raised by the gonadotoxicity of chemotherapeutic agents, and some progress has been achieved in the field of fertility preservation in female patients. For example, fertility preservation technologies such as oocyte cryopreservation and embryo cryopreservation have been applied in clinical practice (Ethics Committee of the American Society for Reproductive Medicine, 2018). However, both oocyte cryopreservation and embryo cryopreservation can only be offered to post-pubertal females, while ovarian tissue cryopreservation and transplantation is still an experimental and controversial technology currently with high costs but a low success rate (Oktay and Sönmezer, 2008; Spears et al., 2019). In addition, these fertility preservation technologies are invasive, and their adoption could increase the risk of delaying anti-cancer treatment. Therefore, the development of substances capable of protecting oocytes against the deleterious effects of chemotherapeutic drugs represents a potential major improvement to preserve fertility for these women (Rossi et al., 2017).

cis-Diamminedichloroplatinum (cisplatin) is one of most widely used drugs among the many chemotherapeutic drugs that are widely used for numerous cancers. Although cisplatin is a well-known anti-cancer drug, it is also known for its wide-ranging adverse effects. For example, cisplatin has been proved to be genotoxic in ovarian cells and able to cause loss of follicles in both immature and adult mice (Gonfloni et al., 2009; Nguyen et al., 2019). In fact, the ovarian toxicity of cisplatin is considered second only to that of alkylating agents (Hao et al., 2019). It is therefore urgent to determine the mechanism underlying the ovarian damage caused by cisplatin and develop corresponding protective measures.

Melatonin is an pluripotent endogenous factor shown to have important physiological and pathophysiological roles in the ovary as its receptors are present on ovarian granulosa cells and its receptor-independent antioxidative activity can counter the reactive oxygen species (ROS) that are involved in folliculogenesis, follicular atresia, ovulation, oocyte maturation and corpus luteum formation (Tamura et al., 2009). In addition, melatonin has been put forward as a potential fertoprotective agent in fertility preservation as it can reduce sperm motility in cisplatin-treated testicles and prevent primordial follicle loss in cisplatin-treated ovaries (Jang et al., 2017). However, more detailed investigation is still needed to address some important issues such as whether melatonin has an adverse impact on the anti-cancer effect of cisplatin and what the mechanism of fertility preservation of melatonin in response to cisplatin treatment might be.

The present study was conducted to explore the potential of using melatonin as a fertoprotective adjuvant during cisplatin treatment. Different treatments were performed on BALB/c-nu nude (BALB/c-nu) tumour-bearing mice and tumour-free sexually mature as well as 5-day-old C57BL/6 female mice to illustrate the effect and mechanism of ovarian protection afforded by melatonin in the presence of cisplatin.

Section snippets

Animals

Six-week old athymic BALB/c-nu mice (HFK Bioscience, China), sexually mature female C57BL/6 mice (8 weeks old) and newborn C57BL/6 mice (5 days after birth) were obtained from the Disease Prevention and Control Center of Hubei Province, China. All mice were housed under specific pathogen-free conditions in a temperature-controlled room (22 ± 2°C) with 12 h of light every day. All experimental procedures were performed according to the international ethical guidelines and with the approval of

Melatonin enhances the anti-cancer effect of cisplatin in athymic BALB/c-nu female mice

As the basis of this study, the in-vivo xenograft model was first used to verify the impact of melatonin on the anti-cancer effect of cisplatin. Co-administration of melatonin with cisplatin delayed C13* tumour growth in vivo when compared with the control group or cisplatin mono-therapy or melatonin mono-therapy (Figure 1a), as indicated by the significantly smaller tumour volume in the melatonin + cisplatin group compared with the control group (P < 0.005), melatonin group (P = 0.014) and

Discussion

As routine side effects of anti-cancer therapies, premature ovarian failure and infertility are frequent in young cancer survivors owing to the extreme sensitivity of ovarian follicles to the damaging effects of chemotherapeutic drugs (Oktem and Oktay, 2007). It is of great importance and urgency that agents should be found that will be suitable for patients of all ages and life stages to prevent follicle depletion at the time of anti-cancer treatment.

The fact that ovarian cancer-bearing

Acknowledgements

This study was supported by the National Key Research and Development Program (2019YFC1005200, 2019YFC1005202), National Natural Science Foundation of China (81402163, 81572569, 81802896), Natural Science Foundation of Hubei Province (2017CFB800), Hubei Province Health and Family Planning Scientific Research Project (WJ2017Z013, WJ2019M127) and National Key Research and Development Program (2018YFC1002103).

Professor Kezhen Li, MD, PhD, has worked at Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China since 2003. She devotes herself to both basic and clinical research into oncofertility and gynaecological oncology.

Key message

Co-administration of melatonin with cisplatin improved control of tumour growth, ovarian reserve and fertility in female mice. Mechanistically, melatonin reduced DNA damage and alleviated lipid peroxidation toxicity

References (38)

  • C. Borgeest et al.

    Methoxychlor may cause ovarian follicular atresia and proliferation of the ovarian epithelium in the mouse

    Toxicol. Sci.

    (2002)
  • F.J. Broekmans et al.

    Ovarian aging: mechanisms and clinical consequences

    Endocr. Rev.

    (2009)
  • M.C. Cora et al.

    Vaginal Cytology of the Laboratory Rat and Mouse: Review and Criteria for the Staging of the Estrous Cycle Using Stained Vaginal Smears

    Toxicol. Pathol.

    (2015)
  • J.A. Flaws et al.

    Chronically elevated luteinizing hormone depletes primordial follicles in the mouse ovary

    Biol. Reprod.

    (1997)
  • S. Gonfloni et al.

    Inhibition of the c-Abl-TAp63 pathway protects mouse oocytes from chemotherapy-induced death

    Nat. Med.

    (2009)
  • X. Hao et al.

    Ovarian Follicle Depletion Induced by Chemotherapy and the Investigational Stages of Potential Fertility-Protective Treatments-A Review

    Int. J. Mol. Sci.

    (2019)
  • X. Hao et al.

    Ovarian Follicle Depletion Induced by Chemotherapy and the Investigational Stages of Potential Fertility-Protective Treatments-A Review

    Int. J. Mol. Sci.

    (2019)
  • N. Jancar et al.

    Effect of apoptosis and reactive oxygen species production in human granulosa cells on oocyte fertilization and blastocyst development

    J. Assist. Reprod. Genet.

    (2007)
  • H. Jang et al.

    Melatonin and Fertoprotective Adjuvants: Prevention against Premature Ovarian Failure during Chemotherapy

    Int. J. Mol. Sci.

    (2017)
  • Cited by (26)

    • New insights into the reverse of chromium-induced reprotoxicity of pregnant mice by melatonin

      2022, Ecotoxicology and Environmental Safety
      Citation Excerpt :

      The daily water intake of pregnant mice in Ctrl and Cr(VI) groups were tracked for four days in Table S1. The MLT dosage was based on previous research (de Souza et al., 2018; Huang et al., 2021), which demonstrated positive responses to reproduction of pregnant mice. All treatments began at GD7 an progressed to GD 19.

    • Therapeutic Effects of Low-Intensity Pulsed Ultrasound on Premature Ovarian Insufficiency

      2021, Ultrasound in Medicine and Biology
      Citation Excerpt :

      Thus, after LIPUS treatment, compared with the control group, the ultrasound group exhibited no obvious improvement AMH secretion, pregnancy rate and fertility status. Most studies on the efficacy of POI treatment have found that fertility can be restored to some extent (Yoon et al. 2020; Huang et al. 2021). Thus, we need to further improve the experimental design, such as appropriately extending the observation time and increasing sample size.

    • Melatonin protected against kidney impairment induced by 5-fluorouracil in mice

      2023, Journal of Experimental Zoology Part A: Ecological and Integrative Physiology
    View all citing articles on Scopus

    Professor Kezhen Li, MD, PhD, has worked at Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China since 2003. She devotes herself to both basic and clinical research into oncofertility and gynaecological oncology.

    Key message

    Co-administration of melatonin with cisplatin improved control of tumour growth, ovarian reserve and fertility in female mice. Mechanistically, melatonin reduced DNA damage and alleviated lipid peroxidation toxicity caused by cisplatin in the ovary. These results indicate that melatonin may be a promising fertoprotective agent for cisplatin-treated female cancer patients.

    #

    These authors should be regarded as joint first authors.

    View full text