1
|
Liu L, Wang H, Wen W, Wang S, Zuo L, Cheng Y, Rao M, Ma Y, Tang L. Humanin alone and in combination with GnRHa therapy attenuates ovarian dysfunction induced by prepubertal cyclophosphamide chemotherapy in female mice. Reprod Toxicol 2025; 132:108824. [PMID: 39793741 DOI: 10.1016/j.reprotox.2024.108824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/01/2024] [Accepted: 12/18/2024] [Indexed: 01/13/2025]
Abstract
Prepubertal chemotherapy induced ovarian damage poses a significant threat to female fertility, particularly following cyclophosphamide (CP) treatment. Humanin (HNG), a small molecule polypeptide encoded by mitochondrial DNA, has a variety of effects, this study aimed to investigate the protective effects of HNG and its combination with conventional Gonadotropin Releasing Hormone Agonist (GnRHa) on ovarian function in a CP-induced damage model. The 21-day-old C57BL/6 J female mice were randomly assigned to six groups: Control, CP model, HNG, HNG+CP, GnRHa+CP, and HNG+GnRHa+CP. Reproductive related parameters were assessed through histopathological examination, follicle counts, serum sex hormone levels, estrous cycle monitoring, and oxidative stress evaluation. Results indicated that CP treatment led to significant reproductive dysfunction especially ovarian dysfunction, evidenced by reduced follicles, hormonal imbalances, prolonged estrous cycles, reduced body weight, and diminished ovarian and uterine weights, alongside pathological alterations. Notably, HNG treatment, both alone and in conjunction with GnRHa, significantly mitigated these adverse effects, however the combination did not provide additional benefits over HNG alone regarding follicles preservation and antioxidant capacity. Transcriptomic analysis revealed significant enrichment in inflammation and immune response pathways following HNG treatment. In conclusion, HNG demonstrates potential as a therapeutic agent to protect against CP-induced ovarian damage, offering insights for future strategies aimed at preserving female fertility during chemotherapy.
Collapse
Affiliation(s)
- Liu Liu
- Reproductive Genetics Department, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Huawei Wang
- Reproductive Genetics Department, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Wen Wen
- Reproductive Genetics Department, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Shunqing Wang
- Reproductive Genetics Department, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Liqin Zuo
- Reproductive Genetics Department, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Yulin Cheng
- Reproductive Genetics Department, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Meng Rao
- Reproductive Genetics Department, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China.
| | - Yuru Ma
- Reproductive Genetics Department, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China.
| | - Li Tang
- Reproductive Genetics Department, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China.
| |
Collapse
|
2
|
Huang Y, Zhang Q, Shen D, Bao X. Mechanisms of He Shi Yu Lin formula in treating premature ovarian insufficiency: insights from network pharmacology and animal experiments. J Ovarian Res 2024; 17:254. [PMID: 39731132 DOI: 10.1186/s13048-024-01575-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 12/08/2024] [Indexed: 12/29/2024] Open
Abstract
OBJECTIVE He Shi Yu Lin Formula (HSYLF) is a clinically proven prescription for treating premature ovarian insufficiency (POI), and has shown a good curative effect. However, its molecular mechanisms are unclear. This study aimed to investigate the molecular mechanisms of HSYLF and clarify how network pharmacology analysis guides the design of animal experiments, including the selection of effective treatment doses and key targets, to ensure the relevance of the experimental results. METHODS Network pharmacology, molecular docking, and animal experiments were utilized to investigate the effects of HSYLF. Key targets were identified by intersecting herb and disease targets to construct protein-protein interaction and "active components-intersection targets-disease" networks. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses were performed using the clusterProfiler package in R. A total of 50 specific pathogen-free female mice of reproductive age were included in the animal experiments. They were divided into five groups: the positive control group, the high-dose HSYLF group, the low-dose HSYLF group, the model blank group, and the normal control group, to evaluate the serum anti-müllerian hormone levels, mitochondrial morphology in oocytes, the levels of reactive oxygen species (ROS), and mitochondrial membrane potential. RESULTS Network pharmacology identified 204 active components connecting 219 key therapeutic targets for POI. Gene Ontology enrichment analysis indicated that the anti-POI targets of HSYLF mainly regulated response to xenobiotic stimulus, cellular response to chemical stress, and response to oxidative stress; and the Kyoto Encyclopedia of Genes and Genomes pathway analysis suggested the primary pathways, including lipid and atherosclerosis, advanced glycation end product-receptor for advanced glycation end product signaling pathway in diabetic complications, bladder cancer, tumor necrosis factor signaling pathway, and interleukin-17 signaling pathway. The low-dose (33 g/kg/d) HSYLF and high-dose (66 g/kg/d) HSYLF groups exhibited a marked elevation in serum anti-müllerian hormone levels (low-dose group: 2657.63 ± 354.82 PG/ml; high-dose group: 2823.73 ± 316.04 PG/ml) and mitochondrial membrane potential compared to the model blank group (P < 0.05 or P < 0.01), along with a significant decline in fluorescence intensity of 2',7'-dichlorofluorescein for the levels of ROS in oocytes (P < 0.05 or P < 0.01). Additionally, both groups showed varying degrees of improvement in the morphology, quantity, and distribution of mitochondria. CONCLUSION This study provides definite evidence for the molecular mechanism by which HSYLF treats POI by decreasing mitochondrial ROS, increasing membrane potential, and improving mitochondrial function. The results from active components of HSYLF and their related key targets also confirmed the characteristics of its multi-component, multi-target, multi-pathway, and overall regulatory effects on POI. Further research regarding the mechanisms is required to generalize these results, and the deeper clinical value of HSYLF also needs to be investigated in the future.
Collapse
Affiliation(s)
- Yun Huang
- TCM Gynecology Department, Hangzhou Hospital of Traditional Chinese Medicine, NO.453 Ti Yuchang Road, Hangzhou, 310007, Zhejiang, China.
- Geriatric Department, Hangzhou Hospital of Traditional Chinese Medicine, NO.453 Ti Yuchang Road, Hangzhou, 310007, Zhejiang, China.
| | - Qin Zhang
- TCM Gynecology Department, Hangzhou Hospital of Traditional Chinese Medicine, NO.453 Ti Yuchang Road, Hangzhou, 310007, Zhejiang, China
| | - Dan Shen
- TCM Gynecology Department, Hangzhou Hospital of Traditional Chinese Medicine, NO.453 Ti Yuchang Road, Hangzhou, 310007, Zhejiang, China
| | - Xi Bao
- TCM Gynecology Department, Hangzhou Hospital of Traditional Chinese Medicine, NO.453 Ti Yuchang Road, Hangzhou, 310007, Zhejiang, China.
| |
Collapse
|
3
|
Yao Y, Wang B, Yu K, Song J, Wang L, Yang X, Zhang X, Li Y, Ma X. Nur77 ameliorates cyclophosphamide-induced ovarian insufficiency in mice by inhibiting oxidative damage and cell senescence. J Ovarian Res 2024; 17:203. [PMID: 39407305 PMCID: PMC11476119 DOI: 10.1186/s13048-024-01532-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 10/06/2024] [Indexed: 10/19/2024] Open
Abstract
Premature ovarian failure (POF) is among the primary causes of ovarian dysfunction that severely affects women's physical and mental health. The main purpose of this study was to explore the expression level of Nerve growth factor-induced protein B (Nur77/NR4A1) in cyclophosphamide (CTX)-induced POF. We then tested whether Nur77 can exert a protective effect after CTX treatment and investigated the mechanism of Nur77's role during ovarian injury. CTX promotes follicular atresia by inducing redox imbalance, apoptosis, and senescence, thereby causing direct toxicity to gonads. Additionally, CTX decreases ovarian reserve consumption by stimulating the excessive activation of primordial follicles. Nur77 can be stimulated by oxidative stress, DNA damage, metabolism, inflammation, etc. However, its relationship with POF remains unelucidated. We here found that Nur77 is expressed at low levels in POF ovaries. Therefore, Nur77 was identified as a regulator of ovarian injury and follicular development. According to the results, Nur77 overexpression alleviated redox imbalances, reduced cell senescence and apoptosis, and improved follicular reserve. Nur77 protects ovarian function by restoring disordered sex hormone levels and estrus cycles and promoting follicle growth and development at all levels. Moreover, the rapamycin protein kinase (AKT)/mammalian target of the rapamycin (mTOR) is a crucial regulator of the primordial follicle pool and follicular development. A relationship was observed between Nur77 and AKT through string and molecular docking. Experiments confirmed the involvement of the AKT/mTOR signaling pathway in the regulatory role of Nur77 in ovarian function. Thus, Nur77 is a critical target for POF prevention and treatment.
Collapse
Affiliation(s)
- Ying Yao
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Bin Wang
- Reproductive Medicine Center, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory for Reproductive Medicine and Embryo of Gansu, Lanzhou, China
| | - Kaihua Yu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Ji Song
- Reproductive Medicine Center, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory for Reproductive Medicine and Embryo of Gansu, Lanzhou, China
| | - Liyan Wang
- Reproductive Medicine Center, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory for Reproductive Medicine and Embryo of Gansu, Lanzhou, China
| | - Xia Yang
- Reproductive Medicine Center, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory for Reproductive Medicine and Embryo of Gansu, Lanzhou, China
| | - Xuehong Zhang
- Reproductive Medicine Center, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory for Reproductive Medicine and Embryo of Gansu, Lanzhou, China
| | - Yulan Li
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, China.
- , No. 1, Donggang West Road, Chengguan District, Lanzhou City, Gansu Province, China.
| | - Xiaoling Ma
- Reproductive Medicine Center, The First Hospital of Lanzhou University, Lanzhou, China.
- Key Laboratory for Reproductive Medicine and Embryo of Gansu, Lanzhou, China.
- , No. 1, Donggang West Road, Chengguan District, Lanzhou City, Gansu Province, China.
| |
Collapse
|
4
|
Zidan A, Elnady M, Khalifa BN. Donepezil protects against cyclophosphamide-induced premature ovarian failure in mice: A focus on proinflammatory cytokines and NLRP3/TLR-4/NF-κB interplay. Toxicol Appl Pharmacol 2024; 488:116989. [PMID: 38825044 DOI: 10.1016/j.taap.2024.116989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 06/04/2024]
Abstract
BACKGROUND AND AIM Cyclophosphamide (CP) chemotherapy is a significant iatrogenic component of premature ovarian failure (POF). The aim of this work was to evaluate the potential protective effects of donepezil, a centrally acting acetylcholinesterase (AChE) inhibitor, on CP-induced POF in mice. METHODS 40 female Swiss albino mice were split into 5 equal groups: group 1 (control), group 2 (CP-POF); induced by intraperitoneal injection of CP on 8th day of the experiment, and group (3-5); mice received oral donepezil daily (1, 2, or 4 mg/kg, respectively) 8 days before CP injection. Mice were euthanized after 24 h of CP injection, and blood samples were collected to assay serum anti-Mullerian hormone (AMH) levels. Ovarian tissues were dissected, and the right ovary was processed for further assays of nitric oxide (NO), tumor necrosis factor-α (TNF-α), interlukin-6 (IL-6), nucleotide-binding domain-like receptor family, the Pyrin domain-containing 3 (NLRP3) inflammasome, and Toll-like receptor 4 (TLR-4), while the left one was processed for histopathological and immunohistochemical examination of nuclear factor-Kappa beta (NF-κB) and caspase-3. RESULTS Donepezil, in a dose-dependent manner particularly (4 mg/kg), has an inhibitory action on NO (40 ± 2.85 vs. 28.20 ± 2.23, P < 0.001), proinflammatory cytokines (P < 0.001), the TLR-4/ NF-κB / NLRP3 inflammasome pathway (P < 0.001), and apoptosis (P < 0.001), with a significant elevation in the AMH levels (4.57 ± 1.08 vs. 8.57 ± 0.97, P < 0.001) versus CP-POF group. CONCLUSION Donepezil may be a potential protective agent against CP-induced POF in mice, but further research is needed to fully understand its therapeutic function experimentally and clinically.
Collapse
Affiliation(s)
- Amr Zidan
- Department of Pharmacology, Faculty of medicine, Tanta University, Egypt.
| | - Manar Elnady
- Department of Pathology, Faculty of medicine, Tanta University, Egypt
| | - Basma N Khalifa
- Department of Pharmacology, Faculty of medicine, Tanta University, Egypt
| |
Collapse
|
5
|
Markowska A, Antoszczak M, Markowska J, Huczyński A. Gynotoxic Effects of Chemotherapy and Potential Protective Mechanisms. Cancers (Basel) 2024; 16:2288. [PMID: 38927992 PMCID: PMC11202309 DOI: 10.3390/cancers16122288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Chemotherapy is one of the leading cancer treatments. Unfortunately, its use can contribute to several side effects, including gynotoxic effects in women. Ovarian reserve suppression and estrogen deficiency result in reduced quality of life for cancer patients and are frequently the cause of infertility and early menopause. Classic alkylating cytostatics are among the most toxic chemotherapeutics in this regard. They cause DNA damage in ovarian follicles and the cells they contain, and they can also induce oxidative stress or affect numerous signaling pathways. In vitro tests, animal models, and a few studies among women have investigated the effects of various agents on the protection of the ovarian reserve during classic chemotherapy. In this review article, we focused on the possible beneficial effects of selected hormones (anti-Müllerian hormone, ghrelin, luteinizing hormone, melatonin), agents affecting the activity of apoptotic pathways and modulating gene expression (C1P, S1P, microRNA), and several natural (quercetin, rapamycin, resveratrol) and synthetic compounds (bortezomib, dexrazoxane, goserelin, gonadoliberin analogs, imatinib, metformin, tamoxifen) in preventing gynotoxic effects induced by commonly used cytostatics. The presented line of research appears to provide a promising strategy for protecting and/or improving the ovarian reserve in the studied group of cancer patients. However, well-designed clinical trials are needed to unequivocally assess the effects of these agents on improving hormonal function and fertility in women treated with ovotoxic anticancer drugs.
Collapse
Affiliation(s)
- Anna Markowska
- Department of Perinatology and Women’s Health, Poznań University of Medical Sciences, 60-535 Poznań, Poland
| | - Michał Antoszczak
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, 61-614 Poznań, Poland
| | - Janina Markowska
- Gynecological Oncology Center, Poznańska 58A, 60-850 Poznań, Poland;
| | - Adam Huczyński
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, 61-614 Poznań, Poland
| |
Collapse
|
6
|
Yu Y, Yang K. Secondary amenorrhea in a β-thalassemia major patient treated with thalidomide. Pediatr Blood Cancer 2024; 71:e30982. [PMID: 38526227 DOI: 10.1002/pbc.30982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 03/26/2024]
Affiliation(s)
- Yang Yu
- Department of Clinical Laboratory, Jiang'an County Hospital of Traditional Chinese Medicine, Yibin, China
| | - Kun Yang
- Department of Hematology, Zigong First People's Hospital, Zigong, China
| |
Collapse
|
7
|
Roberts JE, Benoit J, Foong S, Saumet J, Korkidakis A, Marr K, McQuillan S, Todd N. Fertility preservation in patients undergoing gonadotoxic treatments: a Canadian Fertility and Andrology Society clinical practice guideline. Reprod Biomed Online 2024; 48:103767. [PMID: 38458057 DOI: 10.1016/j.rbmo.2023.103767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 10/22/2023] [Accepted: 10/23/2023] [Indexed: 03/10/2024]
Abstract
The management of young patients with cancer presents several unique challenges. In general, these patients are ill prepared for the diagnosis and the impact on their fertility. With the improved survival for all tumour types and stages, the need for adequate fertility counselling and a multidisciplinary approach in the reproductive care of these patients is paramount. Recent advances in cryopreservation techniques allow for the banking of spermatozoa, oocytes, embryos and ovarian tissue without compromising survival. This Canadian Fertility and Andrology Society (CFAS) guideline outlines the current understanding of social and medical issues associated with oncofertility, and the medical and surgical technologies available to optimize future fertility.
Collapse
Affiliation(s)
- Jeffrey E Roberts
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, B.C., Canada.
| | - Janie Benoit
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada
| | - Shu Foong
- Department of Obstetrics and Gynecology, University of Calgary, Calgary, AB, Canada
| | - Julio Saumet
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada
| | - Ann Korkidakis
- Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard University, Boston, MA, USA
| | - Kristin Marr
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, B.C., Canada
| | - Sarah McQuillan
- Department of Obstetrics and Gynecology, University of Calgary, Calgary, AB, Canada
| | - Nicole Todd
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, B.C., Canada
| |
Collapse
|
8
|
Lu G, Li HX, Song ZW, Luo J, Fan YL, Yin YL, Shen J, Shen MH. Combination of bone marrow mesenchymal stem cells and moxibustion restores cyclophosphamide-induced premature ovarian insufficiency by improving mitochondrial function and regulating mitophagy. Stem Cell Res Ther 2024; 15:102. [PMID: 38589967 PMCID: PMC11003045 DOI: 10.1186/s13287-024-03709-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/26/2024] [Indexed: 04/10/2024] Open
Abstract
BACKGROUND Premature ovarian insufficiency (POI) is a major cause of infertility. In this study, we aimed to investigate the effects of the combination of bone marrow mesenchymal stem cells (BMSCs) and moxibustion (BMSCs-MOX) on POI and evaluate the underlying mechanisms. METHODS A POI rat model was established by injecting different doses of cyclophosphamide (Cy). The modeling of POI and the effects of the treatments were assessed by evaluating estrous cycle, serum hormone levels, ovarian weight, ovarian index, and ovarian histopathological analysis. The effects of moxibustion on BMSCs migration were evaluated by tracking DiR-labeled BMSCs and analyzing the expression of chemokines stromal cell-derived factor 1 (Sdf1) and chemokine receptor type 4 (Cxcr4). Mitochondrial function and mitophagy were assessed by measuring the levels of reactive oxygen species (ROS), mitochondrial membrane potential (MMP), ATP, and the mitophagy markers (Drp1, Pink1, and Parkin). Furthermore, the mitophagy inhibitor Mdivi-1 and the mitophagy activator CCCP were used to confirm the role of mitophagy in Cy-induced ovarian injury and the underlying mechanism of combination therapy. RESULTS A suitable rat model of POI was established using Cy injection. Compared to moxibustion or BMSCs transplantation alone, BMSCs-MOX showed improved outcomes, such as reduced estrous cycle disorders, improved ovarian weight and index, normalized serum hormone levels, increased ovarian reserve, and reduced follicle atresia. Moxibustion enhanced Sdf1 and Cxcr4 expression, promoting BMSCs migration. BMSCs-MOX reduced ROS levels; upregulated MMP and ATP levels in ovarian granulosa cells (GCs); and downregulated Drp1, Pink1, and Parkin expression in ovarian tissues. Mdivi-1 significantly mitigated mitochondrial dysfunction in ovarian GCs and improved ovarian function. CCCP inhibited the ability of BMSCs-MOX treatment to regulate mitophagy and ameliorate Cy-induced ovarian injury. CONCLUSIONS Moxibustion enhanced the migration and homing of BMSCs following transplantation and improves their ability to repair ovarian damage. The combination of BMSCs and moxibustion effectively reduced the excessive activation of mitophagy, which helped prevent mitochondrial damage, ultimately improving ovarian function. These findings provide a novel approach for the treatment of pathological ovarian aging and offer new insights into enhancing the efficacy of stem cell therapy for POI patients.
Collapse
Affiliation(s)
- Ge Lu
- College of Acupuncture Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hong-Xiao Li
- College of Acupuncture Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zi-Wei Song
- College of Acupuncture Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jia Luo
- College of Acupuncture Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yan-Liang Fan
- College of Acupuncture Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yao-Li Yin
- College of Acupuncture Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Jie Shen
- College of Acupuncture Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Mei-Hong Shen
- College of Acupuncture Moxibustion and Tuina, Nanjing University of Chinese Medicine, Nanjing, China.
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
9
|
Park SJ, Kim YY, Park W, Park S, Han JY, Kim SW, Kim H, Ku SY. Effects of Cetrorelix on Ovary and Endometrium Prior to Anti-PD-L1 Antibody in Murine Model. Tissue Eng Regen Med 2024; 21:319-328. [PMID: 38103140 PMCID: PMC10825084 DOI: 10.1007/s13770-023-00617-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Recent anti-cancer agents, immune checkpoint inhibitors (ICIs), have emerged as effective agents targeting the programmed cell death protein-1 (PD-1)/programmed death-ligand 1 (PD-L1) pathway. While the administration of gonadotropin-releasing hormone (GnRH) analogs before cytotoxic agents is known to preserve female reproductive organ function, the potential effects of ICIs and the protective impact of GnRH analogs on female reproductive organs, especially concerning ovarian reserve and endometrial receptivity, remain unknown. In this study, we attempted to elucidate the protective or regenerative effect on the female reproductive organ of cetrorelix prior to anti-PD-L1 antibody administration. METHOD Using a murine model, we examined the effects of Anti-PD-L1 antibody treatment on ovarian and uterine morphology, compared them with controls, and further assessed any potential protective effect of cetrorelix, a GnRH analog. Histological examinations and quantitative reverse transcription polymerase chain reaction were employed to study the morphological changes and associated gene expression patterns. RESULTS Anti-PD-L1 treatment led to a significant depletion of primordial/primary ovarian follicles and impaired decidualization in uterine stromal cells. However, while pretreatment with cetrorelix could restore normal decidualization patterns in the uterus, it did not significantly ameliorate ovarian follicular reductions. Gene expression analysis reflected these observations, particularly with marked changes in the expression of key genes like Prl and Igfbp1, pivotal in uterine decidualization. CONCLUSION Our study underscores the potential reproductive implications of cetrorelix treatment prior to Anti-PD-L1 therapy, shedding light on its short-term protective effects on the uterus. Further studies are necessary to understand long-term and clinical implications.
Collapse
Affiliation(s)
- Soo Jin Park
- Department of Obstetrics and Gynecology, Seoul National University Hospital, 101 Daehak-ro Jongno-gu, Seoul, 03080, Republic of Korea
| | - Yoon Young Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, 101 Daehak-ro Jongno-gu, Seoul, 03080, Republic of Korea
| | - Wonhyoung Park
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Sunwoo Park
- Department of Plant & Biomaterials Science, Gyeongsang National University, Jinju-si, 52725, Republic of Korea
- Department of GreenBio Science, Gyeongsang National University, Jinju, 52725, Republic of Korea
| | - Ji Yeon Han
- Department of Obstetrics and Gynecology, Seoul National University Hospital, 101 Daehak-ro Jongno-gu, Seoul, 03080, Republic of Korea
| | - Sung Woo Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, 101 Daehak-ro Jongno-gu, Seoul, 03080, Republic of Korea
| | - Hoon Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, 101 Daehak-ro Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Seung-Yup Ku
- Department of Obstetrics and Gynecology, Seoul National University Hospital, 101 Daehak-ro Jongno-gu, Seoul, 03080, Republic of Korea.
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Republic of Korea.
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
10
|
Feng Y, Zhang W, Xu X, Wang W, Xu Y, Wang M, Zhang J, Xu H, Fu F. Protective effect of Luffa cylindrica fermentation liquid on cyclophosphamide-induced premature ovarian failure in female mice by attenuating oxidative stress, inflammation and apoptosis. J Ovarian Res 2024; 17:24. [PMID: 38273341 PMCID: PMC10809788 DOI: 10.1186/s13048-024-01353-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 01/15/2024] [Indexed: 01/27/2024] Open
Abstract
Premature ovarian failure (POF) is a leading cause of women's infertility without effective treatment. The purpose of this study was to investigate the protective effects of Luffa cylindrica fermentation liquid (LF) on cyclophosphamide (CTX) -induced POF in mice and to preliminarily investigate the underlying mechanisms. Thirty-two Balb/c mice were divided into four groups randomly. One group served as the control, while the other three received CTX injections to establish POF models. A 14-day gavage of either 5 or 10 μL/g LF was administered to two LF pretreatment groups. To analyze the effects of LF, the ovarian index, follicle number, the levels of serum sex hormones, superoxide dismutase (SOD), glutathione (GSH), malondialdehyde (MDA), inflammatory factors, and apoptosis of the ovarian cells were measured. The effects of LF pretreatment on the expression of TLR4/NF-κB and apoptosis pathways were also evaluated. We found that LF pretreatment increased the ovarian index and the number of primordial and antral follicles while decreasing those of atretic follicles. LF pretreatment also increased the serum levels of estradiol (E2) and anti-Müllerian hormone (AMH), while decreasing those of luteinizing hormone (LH) and follicle-stimulating hormone (FSH). Furthermore, LF pretreatment increased the levels of SOD and GSH in the ovaries, while decreasing those of MDA, tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β). LF administration reduced the amount of TUNEL+ ovarian cells and the levels of TLR4 and NF-κB P65 protein expression. In conclusion, LF has antioxidant, anti-inflammatory as well as anti-apoptotic effects against CTX-induced POF, and the inhibition of TLR4/NF-κB and apoptosis pathways may be involved in its mechanisms.
Collapse
Affiliation(s)
- Yueying Feng
- The Second Affiliated Hospital of Nanchang University, Nanchang University, No. 1 Mingde Road, Nanchang, 330000, People's Republic of China
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 Nanjing East Road, Nanchang, 330047, People's Republic of China
| | - Wei Zhang
- The Second Affiliated Hospital of Nanchang University, Nanchang University, No. 1 Mingde Road, Nanchang, 330000, People's Republic of China
| | - Xiaowei Xu
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 Nanjing East Road, Nanchang, 330047, People's Republic of China
| | - Wanzhen Wang
- The Second Affiliated Hospital of Nanchang University, Nanchang University, No. 1 Mingde Road, Nanchang, 330000, People's Republic of China
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 Nanjing East Road, Nanchang, 330047, People's Republic of China
| | - Yuanyuan Xu
- The Second Affiliated Hospital of Nanchang University, Nanchang University, No. 1 Mingde Road, Nanchang, 330000, People's Republic of China
| | - Mengqi Wang
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 Nanjing East Road, Nanchang, 330047, People's Republic of China
| | - Jinfeng Zhang
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 Nanjing East Road, Nanchang, 330047, People's Republic of China
| | - Hengyi Xu
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 Nanjing East Road, Nanchang, 330047, People's Republic of China.
| | - Fen Fu
- The Second Affiliated Hospital of Nanchang University, Nanchang University, No. 1 Mingde Road, Nanchang, 330000, People's Republic of China.
| |
Collapse
|
11
|
Kashi O, Meirow D. Overactivation or Apoptosis: Which Mechanisms Affect Chemotherapy-Induced Ovarian Reserve Depletion? Int J Mol Sci 2023; 24:16291. [PMID: 38003481 PMCID: PMC10671775 DOI: 10.3390/ijms242216291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Dormant primordial follicles (PMF), which constitute the ovarian reserve, are recruited continuously into the cohort of growing follicles in the ovary throughout female reproductive life. Gonadotoxic chemotherapy was shown to diminish the ovarian reserve pool, to destroy growing follicle population, and to cause premature ovarian insufficiency (POI). Three primary mechanisms have been proposed to account for this chemotherapy-induced PMF depletion: either indirectly via over-recruitment of PMF, by stromal damage, or through direct toxicity effects on PMF. Preventative pharmacological agents intervening in these ovotoxic mechanisms may be ideal candidates for fertility preservation (FP). This manuscript reviews the mechanisms that disrupt follicle dormancy causing depletion of the ovarian reserve. It describes the most widely studied experimental inhibitors that have been deployed in attempts to counteract these affects and prevent follicle depletion.
Collapse
Affiliation(s)
- Oren Kashi
- The Morris Kahn Fertility Preservation Center, Sheba Medical Center, Ramat Gan 5262000, Israel;
| | - Dror Meirow
- The Morris Kahn Fertility Preservation Center, Sheba Medical Center, Ramat Gan 5262000, Israel;
- Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
12
|
Xiu Z, Tang S, Kong P, Yan M, Tong X, Liu X, Liang X, Li R, Duan Y. The effect and mechanism of Zigui-Yichong-Fang on improving ovarian reserve in premature ovarian insufficiency by activating SIRT1/Foxo3a pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023; 314:116608. [PMID: 37150421 DOI: 10.1016/j.jep.2023.116608] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/24/2023] [Accepted: 05/04/2023] [Indexed: 05/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Zigui-Yichong-Fang (ZGYCF) is a traditional Chinese medicine prescription for the treatment of infertility. It is clinically used to regulate the hormone level of patients, improve ovarian reserve function and increase pregnancy rate. However, the exact mechanism of action is not yet clear. AIMS OF THE STUDY This study aims to explore the potential impact of ZGYCF on POI and its mechanism. MATERIALS AND METHODS Ultra-high performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) was used to identify the main compounds of ZGYCF. After confirming the therapeutic effect of ZGYCF on cyclophosphamide-induced POI mice, RNA sequencing (RNA-seq) analysis was carried out to explore the mechanism. Then, the effects of ZGYCF on SIRT1 deacetylated Foxo3a and apoptosis were verified from multiple perspectives by serum hormone level, mRNA validation, histomorphology and protein expression, acetylation modification and other experiments. RESULTS ZGYCF can improve the morphological changes of ovarian tissue in POI model mice, reduce the damage of primordial follicles and other follicles at all stages, and protect ovarian reserve. The results of transcriptome sequencing showed that the genes expression of PI3K signal and apoptosis signal pathway were increased in POI model mice; ZGYCF can up-regulate the expression of SIRT1 gene and the expression of estradiol, apoptosis inhibition and other signal pathway genes. In addition, ZGYCF can reduce follicular damage and ovarian cell apoptosis in POI model mice through activating the deacetylation of Foxo3a by SIRT1, and improve ovarian reserve function. CONCLUSIONS ZGYCF may improve ovarian reserve function of CTX-induced POI mice by activating SIRT1-mediated deacetylation of Foxo3a, and play a role in the treatment of POI.
Collapse
Affiliation(s)
- Zi Xiu
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China
| | - Siling Tang
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China
| | - Peng Kong
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Mengxuan Yan
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China
| | - Xue Tong
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China
| | - Xueping Liu
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China
| | - Xiao Liang
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China
| | - Rongxia Li
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China
| | - Yancang Duan
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050200, China.
| |
Collapse
|
13
|
Liu B, Liu Y, Li S, Chen P, Zhang J, Feng L. BDNF promotes mouse follicular development and reverses ovarian aging by promoting cell proliferation. J Ovarian Res 2023; 16:83. [PMID: 37106468 PMCID: PMC10134588 DOI: 10.1186/s13048-023-01163-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 04/16/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND Brain-derived neurotrophic factor (BDNF) plays an important role in ovarian function including follicle development and oocyte maturation, and embryonic development. However, whether BDNF treatment can reimpose ovarian aging and impaired fertility remains elusive. In this study, we investigated the reproductive outcomes of BDNF treatment and potential mechanisms in aged mice. METHOD "Aged" mice (35-37 weeks old, n = 68) were treated with recombinant human BDNF protein (rhBDNF, 1 µg/200 µL) through daily intraperitoneal (IP) injection for 10 days with/without ovulation induction. Reproductive age mice (8-10 weeks old, n = 28) were treated with ANA 12 (a selective BDNF receptor, TrkB antagonist) through daily IP injection for 5 days with/without ovulation induction. Ovarian function was assessed by ovarian weight, number of follicles, and sex hormone productions. Following induction of ovulation, the number of total oocytes or abnormal oocytes, and blastocyst formation were assessed. Reproductive functions of the mice were evaluated, including pregnancy rate, mating duration for conception, implantation sites, litter size, and weight of offspring. Finally, the molecular mechanism of the effects of BDNF on ovarian cell functions in mice were examined by Western blot and immunofluorescence. RESULTS rhBDNF treatment increased the ovarian weight, number of follicles, number and quality of oocytes including increased blastocysts formation, blood estrogen levels, and pregnancy rate in 35-37-week-old mice. Conversely, BDNF receptor antagonist, ANA 12, treatment decreased the ovarian volume and number of antral follicles and increased the proportion of abnormal oocytes in 8-10-week-old mice. We further demonstrated that BDNF treatment promoted ovarian cell proliferation as well as activation of TrkB and cyclinD1-creb signalling. CONCLUSION We demonstrated that ten consecutive days of daily IP injection of rhBDNF rescued ovarian function in aged mice. Our results further indicate that TrkB and cyclin D1-creb signaling may underlie the BDNF function in ovaries. Targeting BDNF-TrkB signaling is a potential novel therapeutic strategy to reverse ovarian aging.
Collapse
Affiliation(s)
- Bin Liu
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Reproduction, Xinhua Hospital, School of Medicine, Shanghai Jiao-Tong University, Shanghai, China
| | - Yongjie Liu
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shuman Li
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Pingping Chen
- Department of Reproduction, Xinhua Hospital, School of Medicine, Shanghai Jiao-Tong University, Shanghai, China
| | - Jun Zhang
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Liping Feng
- Department of Obstetrics and Gynaecology, Duke University, Durham, NC, USA.
| |
Collapse
|
14
|
Ma H, Wang Y, Liu G, Hu Q, Zhu J, Dai Y. Ovarian scaffolds promoted mouse ovary recovery from cyclophosphamide damage. J Reprod Immunol 2023; 157:103950. [PMID: 37079974 DOI: 10.1016/j.jri.2023.103950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 04/08/2023] [Accepted: 04/12/2023] [Indexed: 04/22/2023]
Abstract
There is growing evidence to suggest that scaffold of tissue can promote the tissue reparation. In this study, we investigate the effects of ovarian scaffolds on the reparation of cyclophosphamide (CPA) damaged mice ovaries. The mice were first administered with CPA, was then either transplanted an ovarian scaffold into each ovarian bursa for the experimental group (EG) or underwent sham surgery as the control (CG). To evaluate the extent of ovarian damage caused by CPA, a third group which did not undergo any treatment was included for the normal control (NG). Their ovaries were harvested for examination at day 30, 60, and 90 post CPA injection. We found that in EG, the number of all types of follicles in the ovaries remained almost the same throughout. The numbers of follicles were not significantly different from CG, except at day 60, where in CG the numbers of each type of follicle decreased to basal levels. The decrease in the number of ovarian follicles at day 60 in CG was mirrored by the significant increase in the number of apoptotic granulosa cells in the follicles, and was corroborated further by the basal levels of serum estradiol. Furthermore, we observed a significant decrease in collagen composition preceded by macrophage polarization, and elevation of inflammatory cytokine expression in the ovaries of the EG compared to the CG at day 60. We concluded that ovarian scaffolds can effectively protect primordial follicles from CPA-damage and promote the reparation of CPA-damaged ovaries. This research establishes a proof of concept for the future treatment of chemo-damaged ovaries.
Collapse
Affiliation(s)
- Hongmeng Ma
- College of Biological Sciences, Inner Mongolia University, 235 West Univ. Road, Hohhot, 010021, Inner Mongolia, China
| | - Yuxing Wang
- College of Biological Sciences, Inner Mongolia University, 235 West Univ. Road, Hohhot, 010021, Inner Mongolia, China
| | - Gang Liu
- Key Laboratory of Medical Cell Biology, Clinical Medicine Research Center, Affiliated Hospital of Inner Mongolia Medical University, 1 Tongdao North Street, Hohhot, 010050, Inner Mongolia, China
| | - Qike Hu
- College of Biological Sciences, Inner Mongolia University, 235 West Univ. Road, Hohhot, 010021, Inner Mongolia, China
| | - Jie Zhu
- College of Biological Sciences, Inner Mongolia University, 235 West Univ. Road, Hohhot, 010021, Inner Mongolia, China
| | - Yanfeng Dai
- College of Biological Sciences, Inner Mongolia University, 235 West Univ. Road, Hohhot, 010021, Inner Mongolia, China.
| |
Collapse
|
15
|
Lee P, Zhou C, Hu X. VX-765 has a Protective Effect on Mice with Ovarian Injury Caused by Chemotherapy. Curr Cancer Drug Targets 2023; 23:307-318. [PMID: 36200259 PMCID: PMC10202083 DOI: 10.2174/1568009622666220930110024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 06/29/2022] [Accepted: 08/15/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Malignant tumors continue to remain a main global public health issue. In the past 40 years, due to strides made in multi-disciplinary comprehensive treatment schemes for patients suffering from malignant tumors, especially chemotherapy schemes, the survival rate has been greatly improved in such patients. This group can be expected to maintain their fertility or have restored endocrine function following successful malignant tumor treatment. Therefore, focusing on the ovarian damage caused by chemotherapy in women of childbearing age is vital in order to protect their fertility and improve their quality of life. OBJECTIVE This study attempted to evaluate whether VX-765 possesses an ovarian protective effect in ovarian injury induced by chemotherapy in the mice model. METHODS Female C57BL/6J mice were administered with VX-765 gavage once a day for 21 consecutive days. Use of cyclophosphamide (Cy) began one week after the last gavage administration of VX- 765. Detailed classification of follicles at various levels was then quantified in each group. Immunohistochemistry and Western blot analysis were then used in order to analyze the expression of key proteins (FOXO3a, mTOR, RPS6 and AKT) as well as their phosphorylation of the PI3K / PTEN / AKT pathways in the ovary. The concentrations of AMH were measured by ELISA. RESULTS The follicles at all levels of Cy treated mice were less than those of the normal group (P < 0.05). Meanwhile, mice treated with VX-765 prior to receiving Cy treatment had more primordial follicles (PMF) than mice treated with Cy alone (P < 0.05). In early growing follicles (EGF) and antral follicles (AF), no difference was observed among the experimental groups (P > 0.05), however, they were lower than those in the normal group (P < 0.05). In mice treated with continuous Cy, the total follicle number (TF) of mice combined with VX-765 (C-Cy-Vx765) was higher than that of mice without VX-765, and the TF of the two groups was lower than that of the normal group (P < 0.05). The value of PMF/TF in C-Cy-Vx765 group was significantly higher than that in the other three groups, while that of EGF/TF was significantly lower (P < 0.05). Immunohistochemical results showed that the phosphorylated forms of the main proteins of the PI3K / PTEN / AKT pathway were found to be more positive in Cy treated mice. The Western blot analysis showed that when Cy and VX-765 were cotreated, the increased levels of these phosphorylated proteins decreased compared with those treated with Cy alone. The AMH level of infancy Cy and VX-765 co-treated mice was higher than that of infancy normal mice (P < 0.05). After the mice grew to sexual maturity, the AMH level of Cy and VX- 765 co-treated mice was still higher than that of Cy treated mice (P < 0.05), and there was no significant difference with normal mice (P > 0.05). CONCLUSION VX-765 can maintain the level of AMH and inhibit the recruitment of PMF, thus protecting mice from Cy induced gonadotropic toxicity. Accordingly, VX-765 may play a protective role in mice with ovarian injury caused by chemotherapy.
Collapse
Affiliation(s)
- Pingyin Lee
- Reproductive Medicine Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Canquan Zhou
- Reproductive Medicine Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaokun Hu
- Reproductive Medicine Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
16
|
Hyperoside protects against cyclophosphamide induced ovarian damage and reduced fertility by suppressing HIF-1α/BNIP3-mediated autophagy. Biomed Pharmacother 2022; 156:113743. [PMID: 36252358 DOI: 10.1016/j.biopha.2022.113743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/03/2022] [Accepted: 09/21/2022] [Indexed: 01/18/2023] Open
|
17
|
Cioffi R, Cervini L, Taccagni G, Papaleo E, Pagliardini L, Bergamini A, Ferrari S, Mangili G, Candiani M. A prospective, observational study of chemotherapy-induced ovarian damage on follicular reserve and maturation. Arch Gynecol Obstet 2022; 306:1723-1729. [PMID: 35833992 DOI: 10.1007/s00404-022-06692-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 06/26/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Chemotherapy negatively affects gonadal function, often resulting in premature ovarian failure (POF) due to ovarian reserve depletion. Mechanisms of gonadotoxicity, such as primordial follicle overactivation and "burnout", remain to be established. Ovarian tissue cryopreservation (OTC) before treatment plays an important role in safeguarding fertility. METHODS This is a prospective observational study that aims to evaluate the feasibility of OTC after chemotherapeutic treatment initiation. Patients were divided into 2 groups depending on whether they received chemotherapy before the harvesting procedure (Group 1) or not (Group 2). The main outcomes of this study are serum anti-Mullerian hormone (AMH) levels and histological follicular counts on ovarian tissue biopsies. RESULTS Between 2012 and 2020, 79 patients underwent OTC at our Hospital. Follicular counts from the ovarian biopsies of 30 post-pubertal patients and respective serum AMH levels were included in the analysis. AMH levels did not significantly differ between the 2 groups (P = 0.70) as well as the number of primordial follicles (P = 0.73). Ovarian biopsies of patients from Group 1 showed a higher number of primary follicles (P = 0.04) and atretic follicles (P = 0.05) with respect to Group 2. CONCLUSIONS In conclusion, OTC appears to be feasible even after the start of chemotherapeutic treatment, since in treated patients, the main ovarian reserve indicators (number of primordial follicles and serum AMH levels) were not significantly reduced compared to untreated patients. The "burnout" theory of chemotherapeutic damage to the ovary seems to be supported by the higher number of primary follicles found in the ovaries of patients who received chemotherapy before OTC.
Collapse
Affiliation(s)
- Raffaella Cioffi
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy.
| | - Laura Cervini
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Gianluca Taccagni
- Pathology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Enrico Papaleo
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Luca Pagliardini
- Division of Genetics and Cell Biology, Reproductive Sciences Laboratory, San Raffaele Scientific Institute, Milan, Italy
| | - Alice Bergamini
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Stefano Ferrari
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Giorgia Mangili
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Massimo Candiani
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| |
Collapse
|
18
|
Guo L, Gao Q, Zhu J, Jin X, Yin H, Liu T. A Docosahexaenoic Acid Derivative ( N-Benzyl Docosahexaenamide) as a Potential Therapeutic Candidate for Treatment of Ovarian Injury in the Mouse Model. Molecules 2022; 27:molecules27092754. [PMID: 35566104 PMCID: PMC9102315 DOI: 10.3390/molecules27092754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/20/2022] [Accepted: 04/23/2022] [Indexed: 02/04/2023] Open
Abstract
Commonly used clinical chemotherapy drugs, such as cyclophosphamide (CTX), may cause injury to the ovaries. Hormone therapies can reduce the ovarian injury risk; however, they do not achieve the desired effect and have obvious side effects. Therefore, it is necessary to find a potential therapeutic candidate for ovarian injury after chemotherapy. N-Benzyl docosahexaenamide (NB-DHA) is a docosahexaenoic acid derivative. It was recently identified as the specific macamide with a high degree of unsaturation in maca (Lepidium meyenii). In this study, the purified NB-DHA was administered intragastrically to the mice with CTX-induced ovarian injury at three dose levels. Blood and tissue samples were collected to assess the regulation of NB-DHA on ovarian function. The results indicated that NB-DHA was effective in improving the disorder of estrous cycle, and the CTX+NB-H group can be recovered to normal levels. NB-DHA also significantly increased the number of primordial follicles, especially in the CTX+NB-M and CTX+NB-H groups. Follicle-stimulating hormone and luteinizing hormone levels in all treatment groups and estradiol levels in the CTX+NB-H group returned to normal. mRNA expression of ovarian development-related genes was positive regulated. The proportion of granulosa cell apoptosis decreased significantly, especially in the CTX+NB-H group. The expression of anti-Müllerian hormone and follicle-stimulating hormone receptor significantly increased in ovarian tissues after NB-DHA treatment. NB-DHA may be a promising agent for treating ovarian injury.
Collapse
Affiliation(s)
- Lirong Guo
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China; (L.G.); (Q.G.); (J.Z.); (X.J.)
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Qing Gao
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China; (L.G.); (Q.G.); (J.Z.); (X.J.)
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jieqiong Zhu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China; (L.G.); (Q.G.); (J.Z.); (X.J.)
| | - Xiaobao Jin
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China; (L.G.); (Q.G.); (J.Z.); (X.J.)
| | - Hui Yin
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China; (L.G.); (Q.G.); (J.Z.); (X.J.)
- Correspondence: (H.Y.); (T.L.)
| | - Tao Liu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China; (L.G.); (Q.G.); (J.Z.); (X.J.)
- Correspondence: (H.Y.); (T.L.)
| |
Collapse
|
19
|
Xue W, Xue F, Jia T, Hao A. Research and experimental verification of the molecular mechanism of berberine in improving premature ovarian failure based on network pharmacology. Bioengineered 2022; 13:9885-9900. [PMID: 35420511 PMCID: PMC9161839 DOI: 10.1080/21655979.2022.2062104] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Based on the research methods of network pharmacology, this study analyzed the improvement effect of berberine (BBR) on premature ovarian failure (POF) and its molecular mechanism. Carry out GO and KEGG enrichment analysis by R language to obtain the potential targets and pathways of BBR in the improvement of POF. Use SD rats and ovarian granulosa cells (GCs) for experimental verification. ELISA was used to measure the content of related hormones in the serum, CCK-8 was used to measure cell viability, western blot was used to measure the content of the target protein in the ovaries and GCs, and q-RT-PCR was used to detect the expression of the target genes in the ovaries and GCs. Predicted by network pharmacology: PTEN, AKT1, FoxO1, FasL, and Bim are the targets with the highest relative correlation between BBR and POF. The results of experiments show that the treatment of low and medium doses of BBR can increase the ovarian index of rats; BBR can increase the levels of Estradiol (E2) and Anti-Mullerian hormone (AMH) in the serum of rats and reduce the levels of Follicle stimulating hormone (FSH) and Luteinizing hormone (LH). BBR can increase the cell viability of GCs; BBR can inhibit the PTEN/AKT1/FoxO1 signaling pathway and its phosphorylation level and reduce the expression of Fas/FasL and Bim mRNA. Overall, BBR can promote the ovarian to maintain normal hormone levels, protect GCs, and enhance the function of POF.
Collapse
Affiliation(s)
- Wu Xue
- Graduate School, Jinzhou Medical University, Jinzhou, Liaoning, China.,Key Laboratory of Follicular Development and Reproductive Health of Liaoning Province, Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - Fan Xue
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Tao Jia
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Ai Hao
- Graduate School, Jinzhou Medical University, Jinzhou, Liaoning, China.,Key Laboratory of Follicular Development and Reproductive Health of Liaoning Province, Jinzhou Medical University, Jinzhou, Liaoning Province, China.,Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| |
Collapse
|
20
|
Mutluay D, Tenekeci GY, Monsef YA. Bortezomib-Induced Ovarian Toxicity in Mice. Toxicol Pathol 2022; 50:381-389. [DOI: 10.1177/01926233221083527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cancer survivors may experience long-term adverse effects of cancer treatments such as premature ovarian failure and infertility. We aimed to investigate the potential effects and toxicity of bortezomib (BTZ) as an effective anticancer drug on ovaries, raise awareness to the negative consequences of the treatment, and help increase the quality of life after treatment. Mice were distributed into bortezomib (BTZ1, BTZ2) and saline-injected control groups (C1, C2) at a dose of 1 mg/kg twice per week for 6 weeks. We sacrificed C1, BTZ1 groups at day 1 and C2, BTZ2 groups at 4 weeks after the last injection. Ovary samples were examined using histopathological and immunohistochemical methods. Ovarian follicle impairment was detected on BTZ-treated mice and was associated with a statistically significant decreased population of primordial and antral follicles compared with control groups. In experimental groups, Caspase-3 and Ki67 expressions were increased, whereas estrogen receptor alpha (ERα) and progesterone receptor (PR) expressions were decreased in various developmental stages of follicles. BTZ specifically targets granulosa cells by inducing granulosa cell apoptosis and may have long-term effects on follicles. Bortezomib treatment may adversely affect ovarian function by accelerating ovarian reserve depletion and changing ERα and PR hormone levels that can cause fertility problems in the long term.
Collapse
Affiliation(s)
- Duygu Mutluay
- Mehmet Akif Ersoy University, Faculty of Veterinary Medicine, Burdur, Turkey
| | | | | |
Collapse
|
21
|
Qin X, Zhao Y, Zhang T, Yin C, Qiao J, Guo W, Lu B. TrkB agonist antibody ameliorates fertility deficits in aged and cyclophosphamide-induced premature ovarian failure model mice. Nat Commun 2022; 13:914. [PMID: 35177657 PMCID: PMC8854395 DOI: 10.1038/s41467-022-28611-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 01/12/2022] [Indexed: 12/18/2022] Open
Abstract
Premature ovarian failure (POF) is a leading cause of women's infertility without effective treatment. Here we show that intravenous injection of Ab4B19, an agonistic antibody for the BDNF receptor TrkB, penetrates into ovarian follicles, activates TrkB signaling, and promotes ovary development. In both natural aging and cyclophosphamide-induced POF models, treatment with Ab4B19 completely reverses the reduction of pre-antral and antral follicles, and normalizes gonadal hormone. Ab4B19 also attenuates gonadotoxicity and inhibits apoptosis in cyclophosphamide-induced POF ovaries. Further, treatment with Ab4B19, but not BDNF, restores the number and quality of oocytes and enhances fertility. In human, BDNF levels are high in granulosa cells and TrkB levels increase in oocytes as they mature. Moreover, BDNF expression is down-regulated in follicles of aged women, and Ab4B19 activates TrkB signaling in human ovary tissue ex vivo. These results identify TrkB as a potential target for POF with differentiated mechanisms, and confirms superiority of TrkB activating antibody over BDNF as therapeutic agents.
Collapse
Affiliation(s)
- Xunsi Qin
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Joint Graduate Program of Peking-Tsinghua-NIBS, Tsinghua University, Beijing, 100084, China
- Beijing Tiantan Hospital, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100070, China
| | - Yue Zhao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Chinese Academy of Medical Sciences, Beijing, 100191, China
| | - Tianyi Zhang
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Joint Graduate Program of Peking-Tsinghua-NIBS, Tsinghua University, Beijing, 100084, China
- Beijing Tiantan Hospital, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100070, China
| | - Chenghong Yin
- Department of Internal Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, 100026, China
| | - Jie Qiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Chinese Academy of Medical Sciences, Beijing, 100191, China
| | - Wei Guo
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Joint Graduate Program of Peking-Tsinghua-NIBS, Tsinghua University, Beijing, 100084, China.
- Beijing Tiantan Hospital, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100070, China.
| | - Bai Lu
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Joint Graduate Program of Peking-Tsinghua-NIBS, Tsinghua University, Beijing, 100084, China.
- Beijing Tiantan Hospital, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100070, China.
| |
Collapse
|
22
|
Lu H, Ma L, Zhang Y, Feng Y, Zhang J, Wang S. Current Animal Model Systems for Ovarian Aging Research. Aging Dis 2022; 13:1183-1195. [PMID: 35855343 PMCID: PMC9286907 DOI: 10.14336/ad.2021.1209] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 12/07/2021] [Indexed: 11/15/2022] Open
Abstract
Ovarian aging leads to menopause, loss of fertility and other disorders in multiple organs, which brings great distress to women. For ethical reasons, it is impossible to use humans as direct study subjects for aging research. Therefore, biomedical researchers have employed different non-human organisms to study ovarian aging, including worms, fruit flies, fishes, amphibians, birds, mice, rats, cavies, rabbits, pigs, sheep, cows, horses, monkeys, and apes. Because each of these model organisms has its own features, multiple factors, such as size, anatomical structure, cost, ease of operation, fertility, generation time, lifespan, and gene heredity, should be carefully considered when selecting a model system to study ovarian aging. An appropriate model organism would help researchers explore the risk factors and elucidate molecular mechanisms underlying declined ovarian functions, which might be conducive to preventing or delaying the ovarian aging process. This article will offer an overview on several currently available and commonly used model organisms for ovarian aging research by comparing their pros and cons. In doing so, we hope to provide useful information for ovarian aging researchers.
Collapse
Affiliation(s)
- Huan Lu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China.
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China.
| | - Lingwei Ma
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China.
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China.
| | - Yan Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China.
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China.
| | - Yanzhi Feng
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China.
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China.
| | - Jinjin Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China.
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China.
- Correspondence should be addressed to: Dr. Shixuan Wang () and Dr. Jinjin Zhang (), Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Wuhan, Hubei, China
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Wuhan, Hubei 430030, China.
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan, Hubei 430030, China.
- Correspondence should be addressed to: Dr. Shixuan Wang () and Dr. Jinjin Zhang (), Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Wuhan, Hubei, China
| |
Collapse
|
23
|
Feng J, Ma WW, Li HX, Pei XY, Deng SL, Jia H, Ma WZ. Melatonin prevents cyclophosphamide-induced primordial follicle loss by inhibiting ovarian granulosa cell apoptosis and maintaining AMH expression. Front Endocrinol (Lausanne) 2022; 13:895095. [PMID: 35992124 PMCID: PMC9381702 DOI: 10.3389/fendo.2022.895095] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Cyclophosphaty -45mide (Cyc) chemotherapy in young female cancer patients is associated with an increased risk of premature ovarian insufficiency (POI). This study was designed to investigate the protective role of melatonin (Mel) as an adjuvant against Cyc-induced POI. Female mice received a single intraperitoneal (i.p.) dose of Cyc (75 mg/kg). Mel protection was achieved in mice after i.p. injection of melatonin (50 mg/kg) every 24 h for four consecutive days prior to chemotherapy initiation and for 14 additional days. Ovarian reserve testing, hormonal assays for follicle-stimulating hormone, luteinizing hormone, and anti-Müllerian hormone (AMH), assessment of the oxidative stress status, and measurement of the relative expression of genes in PTEN/AKT/FOXO3a and mitochondrial apoptosis pathways were performed. The results showed that treatment with 50 mg/kg Mel significantly prevented Cyc-induced over-activation of primordial follicles by maintaining the plasma level of AMH and subsequently preventing litter size reduction in mice treated with Cyc chemotherapy. Importantly, Mel treatment significantly prevented ovarian granulosa cell loss by inhibiting the mitochondrial apoptotic pathway. Identifying the protective actions of Mel against Cyc-induced primordial follicle loss has important implications for fertility maintenance in young cancer patients undergoing chemotherapy.
Collapse
Affiliation(s)
- Juan Feng
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, and Key Laboratory of Reproduction and Genetics of Ningxia Hui Autonomous Region, School of Basic Medical Science, Ningxia Medical University, Yinchuan, China
| | - Wen-Wen Ma
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, and Key Laboratory of Reproduction and Genetics of Ningxia Hui Autonomous Region, School of Basic Medical Science, Ningxia Medical University, Yinchuan, China
| | - Hui-Xia Li
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, and Key Laboratory of Reproduction and Genetics of Ningxia Hui Autonomous Region, School of Basic Medical Science, Ningxia Medical University, Yinchuan, China
| | - Xiu-Ying Pei
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, and Key Laboratory of Reproduction and Genetics of Ningxia Hui Autonomous Region, School of Basic Medical Science, Ningxia Medical University, Yinchuan, China
| | - Shou-Long Deng
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
- *Correspondence: Shou-Long Deng, ; Hua Jia, ; Wen-Zhi Ma,
| | - Hua Jia
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, and Key Laboratory of Reproduction and Genetics of Ningxia Hui Autonomous Region, School of Basic Medical Science, Ningxia Medical University, Yinchuan, China
- *Correspondence: Shou-Long Deng, ; Hua Jia, ; Wen-Zhi Ma,
| | - Wen-Zhi Ma
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, and Key Laboratory of Reproduction and Genetics of Ningxia Hui Autonomous Region, School of Basic Medical Science, Ningxia Medical University, Yinchuan, China
- *Correspondence: Shou-Long Deng, ; Hua Jia, ; Wen-Zhi Ma,
| |
Collapse
|
24
|
Ge E, Zha R, Gao Q, Guo L, Wang B, Lin Q, Zhang G, Xie W, Yin H, Liu T. Preventive effect of swim bladder hydrolysates on cyclophosphamide-induced ovarian injury in mice. J Obstet Gynaecol Res 2021; 48:420-430. [PMID: 34852403 DOI: 10.1111/jog.15122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 10/11/2021] [Accepted: 11/25/2021] [Indexed: 10/19/2022]
Abstract
AIMS This study aimed to prepare swim bladder hydrolysate (SBH) with Mn < 4000 Da, and investigate its effects on cyclophosphamide (CTX)-mediated ovarian injury in mice. METHODS Hydrolysates were prepared by heating extraction, enzymatic hydrolysis and ultrafiltration. Mn and distribution of SBH were analyzed via gel filtration chromatography and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. Changes in the mouse oestrus cycle were determined by cytological examination. The number of follicles was examined using histopathology. Enzyme-linked immunosorbent assays (ELISAs) were used to determine the serum sex hormone levels. RESULTS The Mn of SBH, prepared by heating extraction, enzymatic hydrolysis, ultrafiltration, and from different batches, was below 4000 Da, and the preparation process was stable. Compared with the control group, the low-, middle-, and high-dose SBH treatment groups showed different trends in oestrus duration, serum sex hormone levels, and the number of primordial and secondary follicles. The oestrus cycle duration of the high-dose SBH group was longer than that of the model group. The serum luteinizing hormone, follicle-stimulating hormone, and anti-Müllerian hormone levels in the middle-dose group were the closest to those of control group. The number of primordial and secondary follicles in the medium-dose group was significantly higher than that in the model group and closest to those of control group. CONCLUSION After heating extraction, trypsin/Flavourzyme hydrolysis and ultrafiltration, a hydrolysate with Mn below 4000 Da could be prepared. We found that a moderate (400 mg/kg) SBH dose resulted in the greatest effect on ovarian injury remission in mice.
Collapse
Affiliation(s)
- Enhui Ge
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Rui Zha
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Qing Gao
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China.,School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Lirong Guo
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China.,School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Binglong Wang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Qiqi Lin
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Guifeng Zhang
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Weiquan Xie
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, China.,School of Pharmacy, Guilin Medical University, Guilin, China
| | - Hui Yin
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Tao Liu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
25
|
Zheng S, Ma M, Chen Y, Li M. Effects of quercetin on ovarian function and regulation of the ovarian PI3K/Akt/FoxO3a signalling pathway and oxidative stress in a rat model of cyclophosphamide-induced premature ovarian failure. Basic Clin Pharmacol Toxicol 2021; 130:240-253. [PMID: 34841658 DOI: 10.1111/bcpt.13696] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 10/17/2021] [Accepted: 11/22/2021] [Indexed: 12/28/2022]
Abstract
To investigate the ability of quercetin to improve ovarian function and inhibit ovarian oxidative stress through the PI3K/Akt/FoxO3a signalling pathway in a rat model of premature ovarian failure (POF), we constructed a POF rat model with cyclophosphamide (CTX) and treated it with quercetin. Haematoxylin and eosin staining (H&E staining) was used to observe the morphological changes of the ovaries. The serum levels of AMH, E2, FSH, SOD, GSH-Px and MDA were determined by enzyme-linked immunosorbent assays. The expression of phosphatidylinositol 3-kinase (PI3K), protein kinase B (Akt), forkhead box O3a (FoxO3a) and their phosphorylated forms AMH, FSH and their receptors in the ovary were detected by western blots. The mRNA expression of PI3K, Akt, FOXO3a, AMH, FSH and their receptors was detected by qRT-PCR. Our results showed that quercetin could significantly increase the expression of AMH, E2, SOD and GSH-Px, upregulate the protein expression of AMH, FSH and its receptor and decrease the expression ratio of phosphorylated PI3K, Akt, FOXO3a and the unphosphorylated forms. Moreover, quercetin inhibited the mRNA expression of PI3K, Akt and FOXO3a. These results suggest that quercetin can restore ovarian function and inhibit oxidative stress by regulating the PI3K/Akt/FoxO3a signalling pathway.
Collapse
Affiliation(s)
- Shaoyan Zheng
- Pharmacy Department, Foshan Women and Children Hospital Affiliated to Southern Medical University, Foshan, China
| | - Mingying Ma
- Pharmacy Department, Nanfang Hospital Affiliated to Southern Medical University, Guangzhou, China
| | - Yanxia Chen
- Pharmacy Department, The First Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Miaoxia Li
- Comprehensive Laboratory, Guangdong Huiqun Chinese Traditional Medicine Co., Ltd, Shantou, China
| |
Collapse
|
26
|
Hao X, Anastácio A, Viñals-Ribé L, Santamaria Lacuesta A, Diakaki C, Alonso de Mena S, Liu K, Rodriguez-Wallberg KA. Follicle Rescue From Prepubertal Ovaries After Recent Treatment With Cyclophosphamide-An Experimental Culture System Using Mice to Achieve Mature Oocytes for Fertility Preservation. Front Oncol 2021; 11:682470. [PMID: 34631518 PMCID: PMC8497963 DOI: 10.3389/fonc.2021.682470] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 09/02/2021] [Indexed: 11/13/2022] Open
Abstract
Ovarian tissue cryopreservation is the only feasible method for fertility preservation in prepubertal girls that will undergo gonadotoxic chemotherapy. To date, the only clinical use of cryopreserved tissue is by a later tissue retransplantation to the patient. Clinical challenges in fertility preservation of very young patients with cancer include time constraints that do not allow to retrieve the tissue for cryopreservation before starting chemotherapy and the preclusion of future ovarian tissue transplantation due to the risk of reintroduction of malignant cells in patients with systemic diseases. To overcome these two challenges, we investigated using an experimental model the feasibility of retrieving secondary follicles from ovaries of prepubertal mice after cyclophosphamide (CPA) treatment in increasing doses of 50, 75, and 100 mg/kg. The follicles were thereafter cultured and matured in vitro. The main outcomes included the efficiency of the method in terms of obtained matured oocytes and the safety of these potentially fertility preservative procedures in terms of analyses of oocyte competence regarding normality of the spindle and chromosome configurations. Our findings demonstrated that it was feasible to isolate and culture secondary follicles and to obtain mature oocytes from prepubertal mice ovaries recently treated with CPA. The efficiency of this method was highly demonstrated in the 100 mg/kg CPA group, with near 90% follicle survival rate after 12 days' culture, similarly to control. Around 80% of the follicles met the criteria to put into maturation, and more than 40% of them achieved metaphase II, with normal spindle and chromosome configurations observed. Suboptimal results were obtained in the 50 and 75 mg/kg CPA groups. These paradoxical findings towards CPA dose might probably reflect a more difficult selection of damaged growing follicles from ovaries recently treated with lower doses of CPA and a hampered ability to identify and discard those with reduced viability for the culture.
Collapse
Affiliation(s)
- Xia Hao
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Laboratory of Translational Fertility Preservation, BioClinicum, Stockholm, Sweden
| | - Amandine Anastácio
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Laboratory of Translational Fertility Preservation, BioClinicum, Stockholm, Sweden
| | - Laia Viñals-Ribé
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Laboratory of Translational Fertility Preservation, BioClinicum, Stockholm, Sweden
| | - Ana Santamaria Lacuesta
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Laboratory of Translational Fertility Preservation, BioClinicum, Stockholm, Sweden
| | - Christina Diakaki
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Laboratory of Translational Fertility Preservation, BioClinicum, Stockholm, Sweden
| | - Sara Alonso de Mena
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Laboratory of Translational Fertility Preservation, BioClinicum, Stockholm, Sweden
| | - Kui Liu
- Shenzhen Key Laboratory of Fertility Regulation, Center of Assisted Reproduction and Embryology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.,Department of Obstetrics and Gynecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong, SAR, China
| | - Kenny A Rodriguez-Wallberg
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden.,Laboratory of Translational Fertility Preservation, BioClinicum, Stockholm, Sweden.,Department of Reproductive Medicine, Division of Gynecology and Reproduction, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
27
|
Christin-Maitre S, Givony M, Albarel F, Bachelot A, Bidet M, Blanc JV, Bouvattier C, Brac de la Perrière A, Catteau-Jonard S, Chevalier N, Carel JC, Coutant R, Donadille B, Duranteau L, El-Khattabi L, Hugon-Rodin J, Houang M, Grynberg M, Kerlan V, Leger J, Misrahi M, Pienkowski C, Plu-Bureau G, Polak M, Reynaud R, Siffroi JP, Sonigo C, Touraine P, Zenaty D. Position statement on the diagnosis and management of premature/primary ovarian insufficiency (except Turner Syndrome). ANNALES D'ENDOCRINOLOGIE 2021; 82:555-571. [PMID: 34508691 DOI: 10.1016/j.ando.2021.09.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Premature ovarian insufficiency (POI) is a rare pathology affecting 1-2% of under-40 year-old women, 1 in 1000 under-30 year-olds and 1 in 10,000 under-20 year-olds. There are multiple etiologies, which can be classified as primary (chromosomal, genetic, auto-immune) and secondary or iatrogenic (surgical, or secondary to chemotherapy and/or radiotherapy). Despite important progress in genetics, more than 60% of cases of primary POI still have no identifiable etiology; these cases are known as idiopathic POI. POI is defined by the association of 1 clinical and 1 biological criterion: primary or secondary amenorrhea or spaniomenorrhea of>4 months with onset before 40 year of age, and elevated follicle-stimulating hormone (FSH)>25IU/L on 2 assays at>4 weeks' interval. Estradiol level is low, and anti-Müllerian hormone (AMH) levels have usually collapsed. Initial etiological work-up comprises auto-immune assessment, karyotype, FMR1 premutation screening and gene-panel study. If all of these are normal, the patient and parents may be offered genome-wide analysis under the "France Génomique" project. The term ovarian insufficiency suggests that the dysfunction is not necessarily definitive. In some cases, ovarian function may fluctuate, and spontaneous pregnancy is possible in around 6% of cases. In confirmed POI, hormone replacement therapy is to be recommended at least up to the physiological menopause age of 51 years. Management in a rare diseases center may be proposed.
Collapse
Affiliation(s)
- Sophie Christin-Maitre
- Sorbonne University, Hôpital St Antoine, Assistance Publique- Hôpitaux de Paris (AP-HP), Paris, France.
| | - Maria Givony
- French National Healthcare Network for Rare Endocrine Diseases (FIRENDO), AP-HP, Paris, France
| | - Frédérique Albarel
- Conception University Hospital, Assistance Publique-Hôpitaux de Marseille (AP-HM), Marseille, France
| | - Anne Bachelot
- Sorbonne University, Hôpital de la Pitié-Salpétrière, AP-HP, Paris, France
| | - Maud Bidet
- Clinique mutualiste La Sagesse, Rennes, France
| | - Jean Victor Blanc
- Sorbonne University, Hôpital St Antoine, Assistance Publique- Hôpitaux de Paris (AP-HP), Paris, France
| | | | | | | | | | | | | | - Bruno Donadille
- Sorbonne University, Hôpital St Antoine, Assistance Publique- Hôpitaux de Paris (AP-HP), Paris, France
| | - Lise Duranteau
- Saclay University, Hôpital du Kremlin-Bicêtre, AP-HP, Paris, France
| | - Laïla El-Khattabi
- Paris-Centre University, Hôpital Cochin Port-Royal, AP-HP, Paris, France
| | | | - Muriel Houang
- Sorbonne University, Hôpital Trousseau, AP-HP, Paris, France
| | - Michaël Grynberg
- Saclay University, Hôpital Antoine Béclère, AP-HP, Clamart, France
| | - Véronique Kerlan
- University of Brest, Centre Hospitalier Régional Universitaire, Brest, France
| | - Juliane Leger
- Paris-Centre University, Hôpital Robert Debré, AP-HP, Paris, France
| | | | | | | | - Michel Polak
- Paris Centre University, Hôpital Necker, AP-HP, Paris, France
| | | | | | - Charlotte Sonigo
- Saclay University, Hôpital Antoine Béclère, AP-HP, Clamart, France
| | - Phillipe Touraine
- Sorbonne University, Hôpital de la Pitié-Salpétrière, AP-HP, Paris, France
| | - Delphine Zenaty
- Paris-Centre University, Hôpital Robert Debré, AP-HP, Paris, France
| |
Collapse
|
28
|
Nouri N, Aghebati-Maleki L, Yousefi M. Adipose-Derived Mesenchymal Stem Cells: A Promising Tool in the Treatment of pre mature ovarian failure. J Reprod Immunol 2021; 147:103363. [PMID: 34450435 DOI: 10.1016/j.jri.2021.103363] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 08/03/2021] [Accepted: 08/15/2021] [Indexed: 12/12/2022]
Abstract
Despite being rare, primary ovarian insufficiency (POI) is a significant cause of infertility and deficiency of ovarian hormone in women. Several health risks are also associated with POI, which include dry eye syndrome, reduced density of bones and enhanced fracture risks, troublesome menopausal symptoms, early development of cardiovascular disease, and psychological effects such as declined cognition, reduced perceived psychological support, anxiety, and depression. Replacing premenopausal levels of ovarian sex steroids through proper hormone replacement therapy could improve the quality of life for POI women and ameliorate related health risks. Herein, POI and its complications, in addition to hormone replacement therapies, which are safe and effective, are discussed. It is proposed that the use of HRT) Hormone replacement therapy (formulations which mimic normal production of ovarian hormones could reduce POI-associated morbidity rates if they are continued by the age 50, which is approximately the natural age of menopause. Particular populations of POI women are also addressed, which include those with enhanced risk of ovarian or breast cancer, those with Turner syndrome, those approaching natural menopause, and those who are breastfeeding. It is generally predicted that stem cell-based therapies would be both safe and effective. In fact, several types of cells have been described as safe, though their effectiveness and therapeutic application are yet to be defined. Several factors exist which could affect the results of treatment, such as cell handling, ex-vivo preparation strategies, variations in tissue of origin, potency, and immunocompatibility. Accordingly, cell types potentially effective in regenerative medicine could be recognized. Notably, products of MSCs from various sources of tissues show different levels of regenerative capabilities. The ultimate focus of the review is on adipose tissue-derive MCSs (ADMSCs), which possess exceptional features such as general availability, great ability to proliferate and differentiate, immunomodulatory capabilities, and low immunogenicity.
Collapse
Affiliation(s)
- Narges Nouri
- Student's Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leili Aghebati-Maleki
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mehdi Yousefi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Science, Tabriz, Iran.
| |
Collapse
|
29
|
Oubiña G, Pascuali N, Scotti L, Bianchi S, May M, Martínez JE, Marchese Ragona C, Higuera J, Abramovich D, Parborell F. Local application of low level laser therapy in mice ameliorates ovarian damage induced by cyclophosphamide. Mol Cell Endocrinol 2021; 531:111318. [PMID: 33989716 DOI: 10.1016/j.mce.2021.111318] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 05/05/2021] [Accepted: 05/07/2021] [Indexed: 01/06/2023]
Abstract
The aim of the present study is to assess whether low level laser therapy (LLLT) can protect ovaries from chemotherapy-induced gonadotoxicity using a mice model of premature ovarian failure induced by cyclophosphamide (CTX). LLLT (64 J/cm2) increased the number of antral follicles whilst decreasing the number of atretic follicles compared to CTX alone. LLLT increased the number of primordial follicles compared with those in the CTX group but they did not differ from those in the control group. LLLT treatment increased the number of AMH-positive follicles compared to CTX alone. LLLT application increased ovarian weight, serum progesterone concentration and P450scc protein levels compared to CTX alone. LLLT reduced the apoptosis in antral follicles and the BAX/BCL-2 ratio compared to CTX alone. Vascular morphology, analysed by CD31 and α-SMA immunostaining, was restored in LLLT-treated ovaries compared to CTX alone. In conclusion, application of LLLT prior to CTX might serve as a promising and novel protocol to preserve female fertility in cancer survivors.
Collapse
Affiliation(s)
- Gonzalo Oubiña
- Instituto de Biología y Medicina Experimental (IByME) - CONICET, Buenos Aires, Argentina
| | - Natalia Pascuali
- Instituto de Biología y Medicina Experimental (IByME) - CONICET, Buenos Aires, Argentina
| | - Leopoldina Scotti
- Instituto de Biología y Medicina Experimental (IByME) - CONICET, Buenos Aires, Argentina
| | - Silvia Bianchi
- Instituto de Biología y Medicina Experimental (IByME) - CONICET, Buenos Aires, Argentina
| | - María May
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | | | | | - Dalhia Abramovich
- Instituto de Biología y Medicina Experimental (IByME) - CONICET, Buenos Aires, Argentina
| | - Fernanda Parborell
- Instituto de Biología y Medicina Experimental (IByME) - CONICET, Buenos Aires, Argentina.
| |
Collapse
|
30
|
Transplantation of cryopreserved ovarian tissue in a series of 285 women: a review of five leading European centers. Fertil Steril 2021; 115:1102-1115. [DOI: 10.1016/j.fertnstert.2021.03.008] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/19/2021] [Accepted: 03/04/2021] [Indexed: 01/18/2023]
|
31
|
Li J, Long H, Cong Y, Gao H, Lyu Q, Yu S, Kuang Y. Quercetin prevents primordial follicle loss via suppression of PI3K/Akt/Foxo3a pathway activation in cyclophosphamide-treated mice. Reprod Biol Endocrinol 2021; 19:63. [PMID: 33892727 PMCID: PMC8063466 DOI: 10.1186/s12958-021-00743-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/11/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Chemotherapy improves the survival rates of patients with various cancers but often causes some adverse effects, including ovarian damage, characterised by a decrease in primordial follicle stockpiles. Recent studies have revealed that chemotherapy may stimulate the PI3K signalling pathway, thereby resulting in accelerated primordial follicle activation and a decreased ovarian reserve. Quercetin is an inhibitor of the PI3K pathway; however, its protective effects against chemotherapy-induced follicle loss in mice have not been established. In this study, the effects of quercetin in a mouse model of cyclophosphamide-induced ovarian dysfunction were investigated. METHODS C57BL/6 female mice were used for the study. Paraffin sections of mouse ovaries (n = 30 mice) were stained with haematoxylin and eosin for differential follicle counts. Apoptosis (n = 5 mice per group) was evaluated by TUNEL assay. Immunohistochemical staining for ki67 and Foxo3a (n = 5 mice per group) was performed to evaluate the activation of primordial follicles. The role of the PI3K signalling pathway in the ovaries (n = 45 mice) was assessed by western blotting. RESULTS Quercetin attenuated the cyclophosphamide-induced reduction in dormant primordial follicles. Analysis of the PI3K/Akt/Foxo3a pathway showed that quercetin decreased the phosphorylation of proteins that stimulate follicle activation in cyclophosphamide-induced ovaries. Furthermore, quercetin prevented cyclophosphamide-induced apoptosis in early growing follicles and early antral follicles, maintained anti-Müllerian hormone levels secreted by these follicles, and preserved the quiescence of the primordial follicle pool, as determined by intranuclear Foxo3a staining. CONCLUSIONS Quercetin attenuates cyclophosphamide-induced follicle loss by preventing the phosphorylation of PI3K/Akt/Foxo3a pathway members and maintaining the anti-Müllerian hormone level through reduced apoptosis in growing follicles. Accordingly, quercetin is expected to improve fertility preservation and the prevention of endocrine-related side effects of chemotherapy.
Collapse
Affiliation(s)
- Jianghui Li
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Zhizaoju Road no. 639, Huangpu District, Shanghai, People's Republic of China
| | - Hui Long
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Zhizaoju Road no. 639, Huangpu District, Shanghai, People's Republic of China
| | - Yanyan Cong
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Zhizaoju Road no. 639, Huangpu District, Shanghai, People's Republic of China
| | - Hongyuan Gao
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Zhizaoju Road no. 639, Huangpu District, Shanghai, People's Republic of China
| | - Qifeng Lyu
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Zhizaoju Road no. 639, Huangpu District, Shanghai, People's Republic of China
| | - Sha Yu
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Zhizaoju Road no. 639, Huangpu District, Shanghai, People's Republic of China.
| | - Yanping Kuang
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Zhizaoju Road no. 639, Huangpu District, Shanghai, People's Republic of China.
| |
Collapse
|
32
|
Shai D, Aviel-Ronen S, Spector I, Raanani H, Shapira M, Gat I, Roness H, Meirow D. Ovaries of patients recently treated with alkylating agent chemotherapy indicate the presence of acute follicle activation, elucidating its role among other proposed mechanisms of follicle loss. Fertil Steril 2021; 115:1239-1249. [PMID: 33485607 DOI: 10.1016/j.fertnstert.2020.11.040] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 11/01/2020] [Accepted: 11/25/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To investigate mechanisms of primordial follicle (PMF) loss in vivo in human ovaries shortly after alkylating agent (AA) chemotherapy. DESIGN Cohort study. SETTING Tertiary university medical center. PATIENT(S) Ninety-six women aged 15-39 years who underwent ovarian tissue cryopreservation for fertility preservation. INTERVENTION(S) Fresh ovarian tissue samples were harvested from women treated with AA (n = 24) or non-AA (n = 24) chemotherapy <6 months after treatment and age-matched untreated women (n = 48). MAIN OUTCOME MEASURE(S) Differential follicle counts, time from chemotherapy exposure, immunostaining for apoptosis (cleaved caspase-3) and FOXO3A on tissue harvested within ultrashort time intervals (4-12 days), collagen (Sirius red) and neovascularization (CD34). RESULT(S) AA-treated ovaries had significant loss of PMFs, and significant increase in absolute numbers of growing follicles compared with untreated control ovaries. The number of growing follicles was inversely correlated with time from chemotherapy. Representative staining for FOXO3A observed decreased nuclear localization in PMF oocytes in AA-treated ovaries removed within the ultrashort time interval compared with untreated ovaries. Neither significant loss of PMFs, increase in growing follicles, nor decrease in nuclear FOXO3A were observed in non-AA-treated ovaries. No increased expression of cleaved caspase-3 was seen in PMFs within the ultrashort time interval after AA or non-AA chemotherapy. Significant stromal fibrosis and neovascularization were observed in AA-treated ovaries only after follicle loss had already occurred (4-6 months). CONCLUSION(S) Follicle activation occurs in vivo in ovaries of patients treated with AA, indicating a pathologic mechanism which may contribute to chemotherapy-induced follicle loss.
Collapse
Affiliation(s)
- Daniel Shai
- Morris Kahn Fertility Preservation Center, Sheba Medical Center, Tel Hashomer
| | - Sarit Aviel-Ronen
- Department of Pathology and Talpiot Medical Leadership Program, Sheba Medical Center, Tel Hashomer; Adelson School of Medicine, Ariel University, Ariel, Israel
| | - Itai Spector
- Morris Kahn Fertility Preservation Center, Sheba Medical Center, Tel Hashomer
| | - Hila Raanani
- Morris Kahn Fertility Preservation Center, Sheba Medical Center, Tel Hashomer
| | - Moran Shapira
- Morris Kahn Fertility Preservation Center, Sheba Medical Center, Tel Hashomer
| | - Itai Gat
- Morris Kahn Fertility Preservation Center, Sheba Medical Center, Tel Hashomer
| | - Hadassa Roness
- Morris Kahn Fertility Preservation Center, Sheba Medical Center, Tel Hashomer
| | - Dror Meirow
- Morris Kahn Fertility Preservation Center, Sheba Medical Center, Tel Hashomer.
| |
Collapse
|
33
|
Upson K, O’Brien KM, Hall JE, Tokar EJ, Baird DD. Cadmium Exposure and Ovarian Reserve in Women Aged 35-49 Years: The Impact on Results From the Creatinine Adjustment Approach Used to Correct for Urinary Dilution. Am J Epidemiol 2021; 190:116-124. [PMID: 32242622 PMCID: PMC7946799 DOI: 10.1093/aje/kwaa037] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 08/19/2019] [Accepted: 10/09/2019] [Indexed: 12/19/2022] Open
Abstract
Cadmium is toxic to the ovaries in animal studies, but its association with diminished ovarian reserve in women is not established. We investigated urinary cadmium, a biomarker of long-term exposure, in relation to diminished ovarian reserve, as indicated by elevated serum follicle-stimulating hormone concentrations (≥10 IU/L), in women aged 35-49 years (unweighted n = 1,681). Using data from the Third National Health and Nutrition Examination Survey (1988-1994), we conducted Poisson regression to estimate adjusted relative risks and 95% confidence intervals. Because the best approach to correcting for urinary dilution in spot samples with creatinine remains controversial, we employed 3 approaches: standardization, covariate adjustment, and covariate-adjusted standardization. Our data suggested a modest association with standardization (highest quartile vs. lowest: relative risk (RR) = 1.3, 95% confidence interval (CI): 0.8, 1.9; P for trend = 0.06) and covariate-adjusted standardization (highest quartile vs. lowest: RR = 1.3, 95% CI: 0.9, 1.9; P for trend = 0.05) and a stronger association with covariate adjustment (highest quartile vs. lowest: RR = 1.8, 95% CI: 1.2, 2.9; P for trend = 0.01). The stronger association with covariate adjustment may reflect bias from conditioning on urinary creatinine, a collider in the hypothesized causal pathway. We conclude that cadmium may contribute to ovarian aging in women and that careful consideration of the creatinine adjustment approach is needed to minimize bias.
Collapse
Affiliation(s)
- Kristen Upson
- Correspondence to Dr. Kristen Upson, Department of Epidemiology and Biostatistics, College of Human Medicine, Michigan State University, 909 Wilson Road, Room B601, East Lansing, MI 48824 (e-mail: )
| | | | | | | | | |
Collapse
|
34
|
Diminished Ovarian Reserve Chemotherapy-Induced Mouse Model: A Tool for the Preclinical Assessment of New Therapies for Ovarian Damage. Reprod Sci 2020; 27:1609-1619. [PMID: 32430713 DOI: 10.1007/s43032-020-00191-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Diminished ovarian reserve (DOR) and primary ovarian insufficiency (POI) are primary factors leading to infertility. However, there is a lack of appropriate animal models of DOR usable for assessing new therapeutic strategies. In this study, we aimed to evaluate whether chemotherapy treatment in mice could reproduce features similar of that observed in women with DOR. Twenty-one Nonobese diabetic/severe combined immunodeficiency (NOD/SCID) female mice were allocated to 3 groups (n = 7/group): control, single dose of vehicle (Dimethyl Sulfoxide [DMSO]); DOR, single reduced chemotherapy dose; and POI, single standard chemotherapy dose. After 21 days, mice underwent ovarian hyperstimulation and mating. Part of the animals were harvested to analyze ovarian reserve, ovulation and fertilization rates, and morphology, apoptosis, and vascularization of the ovarian stroma. The remaining mice underwent multiple matings to assess pregnancy rates and litter sizes. The DOR and POI mice showed an impaired estrous cyclicity and a decrease in ovarian mass, number of follicles, Metaphase II (MII) oocytes, and embryos as well as in ovarian stroma vascularization. Mice in both models showed also an increase in the percentage of morphologically abnormal follicles, stromal degeneration, and apoptosis. Similar to that observed in DOR and POI patients, these impairments were less severe in DOR than in POI mice. None of the POI females were able to achieve a pregnancy. Meanwhile, DOR females achieved several consecutive pregnancies, although litter size was decreased when compared to controls. In conclusion, a mouse model which displayed most of the ovarian characteristics and fertility outcomes of women with DOR has been established using a single dose of chemotherapy.
Collapse
|
35
|
Sarma UC, Winship AL, Hutt KJ. Comparison of methods for quantifying primordial follicles in the mouse ovary. J Ovarian Res 2020; 13:121. [PMID: 33054849 PMCID: PMC7560236 DOI: 10.1186/s13048-020-00724-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/01/2020] [Indexed: 12/29/2022] Open
Abstract
Background Accurate evaluation of primordial follicle numbers in mouse ovaries is an essential endpoint for studies investigating how endogenous and exogenous insults, such as maternal aging and chemotherapy, impact the ovarian reserve. In this study, we compared and contrasted two methods for counting healthy primordial follicles following exposure to cyclophosphamide (75 mg/kg), a well-established model of follicle depletion. The first was the fractionator/optical dissector technique, an unbiased, assumption-free stereological approach for quantification of primordial follicle numbers. While accurate, highly reproducible and sensitive, this method relies on specialist microscopy equipment and software, requires specific fixation, embedding and sectioning parameters to be followed, and is largely a manual process that is tedious and time-consuming. The second method was the more widely used serial section and direct count approach, which is relatively quick and easy. We also compared the impacts of different fixatives, embedding material and section thickness on the overall results for each method. Results Direct counts resulted in primordial follicle numbers that were significantly lower than those obtained by stereology, irrespective of fixation and embedding material. When applied to formalin fixed tissue, the direct count method did not detect differences in follicle numbers between saline and cyclophosphamide treated groups to the same degree of sensitivity as the gold standard stereology method (referred to as the Reference standard). However, when Bouin’s fixative was used, direct counts and stereology were comparable in their ability to detect follicle depletion caused by cyclophosphamide. Conclusions This work indicates that the direct count method can produce similar results to stereology when Bouin’s fixative is used instead of formalin. The findings presented here will assist others to select the most appropriate experimental approach for accurate follicle enumeration, depending on whether the primary objective of the study is to determine absolute primordial follicle numbers or relative differences between groups.
Collapse
Affiliation(s)
- Urooza C Sarma
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Level 3, Building 76, 19 Innovation walk, Clayton, VIC, 3800, Australia.,Department of Anatomy and Developmental Biology, Monash University, Level 3, Building 76, 19 Innovation walk, Clayton, VIC, 3800, Australia
| | - Amy L Winship
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Level 3, Building 76, 19 Innovation walk, Clayton, VIC, 3800, Australia.,Department of Anatomy and Developmental Biology, Monash University, Level 3, Building 76, 19 Innovation walk, Clayton, VIC, 3800, Australia
| | - Karla J Hutt
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Level 3, Building 76, 19 Innovation walk, Clayton, VIC, 3800, Australia. .,Department of Anatomy and Developmental Biology, Monash University, Level 3, Building 76, 19 Innovation walk, Clayton, VIC, 3800, Australia.
| |
Collapse
|
36
|
Li X, Liu S, Ma L, Chen X, Weng H, Huang R, Yu Y, Zong X. Can Anti-Müllerian Hormone Be a Reliable Biomarker for Assessing Ovarian Function in Women Postchemotherapy? Cancer Manag Res 2020; 12:8171-8181. [PMID: 32982414 PMCID: PMC7494398 DOI: 10.2147/cmar.s269249] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/25/2020] [Indexed: 11/23/2022] Open
Abstract
Purpose The predictive value of anti-Müllerian hormone (AMH) for ovarian dysfunction postchemotherapy is controversial. This study aimed to evaluate the value of serum AMH levels clinically and theoretically. Patients, Animals, and Methods We detected the serum estradiol, follicular stimulating hormone (FSH), luteinizing hormone (LH), and AMH levels in 144 premenopausal women with breast cancer receiving cyclophosphamide-based chemotherapy. The hormone levels before and postchemotherapy were compared; the correlations among the hormones and amenorrhea and menstrual recovery were analyzed. In addition, the serum AMH levels were detected randomly in 177 normal healthy women and 36 normal female C57BL/6J mice of different ages; meanwhile, the status of ovarian follicles was also examined. Furthermore, 72 Balb/c nude mice with breast cancer were randomly assigned to three groups that received different doses of cyclophosphamide (CTX) (control, 100 mg/kg, and 200 mg/kg), and the alterations in serum AMH levels and ovarian follicles were recorded and analyzed. Results Chemotherapy-induced amenorrhea was associated with prechemotherapy AMH levels, E2 levels, and FSH levels (P < 0.0001). The recovery of menstruation was associated with prechemotherapy AMH levels (P < 0.0001), but not with E2 and FSH levels (P > 0.05). In patients with breast cancer treated with chemotherapy, the serum AMH levels did not differ significantly between the pre- and post-chemotherapy periods in patients aged <35 years (P > 0.05), whereas a dramatic reduction was detected in patients aged >35 years (P < 0.0001). In healthy women, the serum AMH levels declined sharply after 35 years of age (P < 0.0001) and remained relatively stable at a younger age. Similar results were obtained in experiments using normal mice. The cancer-bearing mice exposed to 200 mg/kg CTX exhibited a significant decline in AMH levels and a remarkable decrease in the number of primordial and growing follicles (P < 0.0001). Conclusion Our results indicate that AMH is an efficient marker for predicting postchemotherapy ovarian function exclusively in premenopausal female patients with breast cancer aged >35 years.
Collapse
Affiliation(s)
- Xiaolin Li
- Department of Breast Surgery, Shanghai Jiao Tong University Affiliated Shanghai Sixth People's Hospital, Shanghai 200233, People's Republic of China.,Institute of Cancer and Basic Medicine, Cancer Hospital of the University of Chinese Academy of Science (Zhejiang Cancer Hospital), Hangzhou 310022, People's Republic of China
| | - Sixuan Liu
- Department of Breast Surgery, Shanghai Jiao Tong University Affiliated Shanghai Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Lisi Ma
- Department of Breast Surgery, Shanghai Jiao Tong University Affiliated Shanghai Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Xuan Chen
- Department of Breast Surgery, Shanghai Jiao Tong University Affiliated Shanghai Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Huaiyu Weng
- Department of Breast Surgery, Shanghai Jiao Tong University Affiliated Shanghai Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Run Huang
- Department of Breast Surgery, Shanghai Jiao Tong University Affiliated Shanghai Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Yang Yu
- Institute of Cancer and Basic Medicine, Cancer Hospital of the University of Chinese Academy of Science (Zhejiang Cancer Hospital), Hangzhou 310022, People's Republic of China
| | - Xiangyun Zong
- Department of Breast Surgery, Shanghai Jiao Tong University Affiliated Shanghai Sixth People's Hospital, Shanghai 200233, People's Republic of China
| |
Collapse
|
37
|
Athira VR, Shivanandappa T, Yajurvedi HN. Cyclophosphamide, a cancer chemotherapy drug-induced early onset of reproductive senescence and alterations in reproductive performance and their prevention in mice. Drug Chem Toxicol 2020; 45:760-766. [DOI: 10.1080/01480545.2020.1774773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
38
|
Al-Kawlani B, Murrieta-Coxca JM, Chaiwangyen W, Fröhlich K, Fritzsche A, Winkler S, Markert UR, Morales-Prieto DM. Doxorubicin induces cytotoxicity and miR-132 expression in granulosa cells. Reprod Toxicol 2020; 96:95-101. [PMID: 32505695 DOI: 10.1016/j.reprotox.2020.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/09/2020] [Accepted: 06/01/2020] [Indexed: 10/24/2022]
Abstract
Doxorubicin (DOX) is one of the most commonly used drugs for the treatment of childhood cancers, including leukemia and lymphomas. Despite the high survival rate, female leukemia survivors are at higher risk of ovarian failure and infertility later in life. Treatment with chemotherapeutic drugs like DOX is associated with damage in ovarian follicles, but the affectation grade of granulosa cells remains unclear. To assess and avoid the possible side-effects of DOX, early biomarkers of ovarian injury and chemotherapy-induced ovarian toxicity should be identified. MicroRNAs (miRNAs) have emerged in recent years as a promising new class of biomarkers for drug-induced tissue toxicity. In this study, the effects of DOX on cell viability, steroidogenesis, and miRNA expression were studied in primary granulosa cells (GCs) and in two cellular models (COV434 and KGN cells). We report that compared to other chemotherapeutic drugs, DOX treatment is more detrimental to granulosa cells as observed by decrease of cell viability. Treatment with DOX changes the expression of the aromatase gene (CYP19A1) and the secretion of 17β-estradiol (E2) in a cell-specific manner. miR-132-3p is dose-dependently increased by DOX in all cellular models. In absence of DOX, miR-132-3p overexpression in COV434 cells has no effect on E2 secretion or CYP19A1 expression. Altogether, these findings contribute to understanding the hormonal disbalance caused by DOX in human ovarian cells and suggest miR-132 as a putative sensor to predict DOX-induced ovarian toxicity.
Collapse
Affiliation(s)
- Boodor Al-Kawlani
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany
| | | | - Wittaya Chaiwangyen
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany; Department of Biochemistry, School of Medical Sciences, University of Phayao, Phayao, Thailand
| | - Karolin Fröhlich
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany
| | | | | | - Udo R Markert
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany.
| | | |
Collapse
|
39
|
Spears N, Lopes F, Stefansdottir A, Rossi V, De Felici M, Anderson RA, Klinger FG. Ovarian damage from chemotherapy and current approaches to its protection. Hum Reprod Update 2020; 25:673-693. [PMID: 31600388 PMCID: PMC6847836 DOI: 10.1093/humupd/dmz027] [Citation(s) in RCA: 369] [Impact Index Per Article: 73.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 07/18/2019] [Accepted: 07/23/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Anti-cancer therapy is often a cause of premature ovarian insufficiency and infertility since the ovarian follicle reserve is extremely sensitive to the effects of chemotherapy and radiotherapy. While oocyte, embryo and ovarian cortex cryopreservation can help some women with cancer-induced infertility achieve pregnancy, the development of effective methods to protect ovarian function during chemotherapy would be a significant advantage. OBJECTIVE AND RATIONALE This paper critically discusses the different damaging effects of the most common chemotherapeutic compounds on the ovary, in particular, the ovarian follicles and the molecular pathways that lead to that damage. The mechanisms through which fertility-protective agents might prevent chemotherapy drug-induced follicle loss are then reviewed. SEARCH METHODS Articles published in English were searched on PubMed up to March 2019 using the following terms: ovary, fertility preservation, chemotherapy, follicle death, adjuvant therapy, cyclophosphamide, cisplatin, doxorubicin. Inclusion and exclusion criteria were applied to the analysis of the protective agents. OUTCOMES Recent studies reveal how chemotherapeutic drugs can affect the different cellular components of the ovary, causing rapid depletion of the ovarian follicular reserve. The three most commonly used drugs, cyclophosphamide, cisplatin and doxorubicin, cause premature ovarian insufficiency by inducing death and/or accelerated activation of primordial follicles and increased atresia of growing follicles. They also cause an increase in damage to blood vessels and the stromal compartment and increment inflammation. In the past 20 years, many compounds have been investigated as potential protective agents to counteract these adverse effects. The interactions of recently described fertility-protective agents with these damage pathways are discussed. WIDER IMPLICATIONS Understanding the mechanisms underlying the action of chemotherapy compounds on the various components of the ovary is essential for the development of efficient and targeted pharmacological therapies that could protect and prolong female fertility. While there are increasing preclinical investigations of potential fertility preserving adjuvants, there remains a lack of approaches that are being developed and tested clinically.
Collapse
Affiliation(s)
- N Spears
- Biomedical Sciences, University of Edinburgh, Edinburgh UK
| | - F Lopes
- Biomedical Sciences, University of Edinburgh, Edinburgh UK
| | | | - V Rossi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - M De Felici
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - R A Anderson
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh UK
| | - F G Klinger
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
40
|
Yoo M, Tanaka T, Konishi H, Tanabe A, Taniguchi K, Komura K, Hayashi M, Ohmichi M. The Protective Effect of Testosterone on the Ovarian Reserve During Cyclophosphamide Treatment. Onco Targets Ther 2020; 13:2987-2995. [PMID: 32308430 PMCID: PMC7152736 DOI: 10.2147/ott.s242703] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/27/2020] [Indexed: 11/23/2022] Open
Abstract
Introduction Cyclophosphamide, which is widely used to treat malignant disease, causes ovarian follicular atresia, which leads to premature ovarian insufficiency. The present study evaluated the protective effect of testosterone in preventing the decline in the ovarian reserve during cyclophosphamide treatment. Methods Using the COV434 human granulosa cell line, the protective effect of testosterone against cyclophosphamide was evaluated by immunocytochemistry, Western blotting and an MTS assay. The follicles in mouse ovaries and serum anti-Mullerian hormone were also assessed to evaluate the effects of testosterone. Results Testosterone suppressed the decrease in cell viability and apoptosis caused by cyclophosphamide treatment in vitro. In vivo, the number of atretic follicles in the mouse ovary was significantly lower in the testosterone plus cyclophosphamide group than in the cyclophosphamide alone group (p=0.03). The serum anti-Mullerian hormone was significantly higher in the testosterone plus cyclophosphamide group than in the cyclophosphamide alone group (16.2 [9.7–22.6]) vs 11.2 [8.9–12.1], p<0.01). The rate of cleaved Caspase-3 expression in the testosterone plus cyclophosphamide group was lower than that in the cyclophosphamide alone group (28.4% vs 48.6%, p=0.03). Conclusion These findings indicated that testosterone has the potential to prevent ovarian damage induced by cyclophosphamide by protecting granulosa cells from cyclophosphamide-induced apoptosis.
Collapse
Affiliation(s)
- Masae Yoo
- Department of Obstetrics and Gynecology
| | - Tomohito Tanaka
- Department of Obstetrics and Gynecology.,Translational Research Program, Osaka Medical College, Takatsuki, Japan
| | | | | | - Kohei Taniguchi
- Translational Research Program, Osaka Medical College, Takatsuki, Japan
| | - Kazumasa Komura
- Translational Research Program, Osaka Medical College, Takatsuki, Japan
| | | | | |
Collapse
|
41
|
Ekiz Yılmaz T, Taşdemir M, Kaya M, Arıcan N, Ahıshalı B. The effects of magnesium sulfate on cyclophosphamide-induced ovarian damage: Folliculogenesis. Acta Histochem 2020; 122:151470. [PMID: 31812447 DOI: 10.1016/j.acthis.2019.151470] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 11/13/2019] [Accepted: 11/13/2019] [Indexed: 01/18/2023]
Abstract
Cyclophosphamide (CYP) is one of the alkylating chemotherapeutic agents and its adverse effects on folliculogenesis in the ovary are well-known due to the previous scientific research on this topic. Magnesium has various effects in organisms, including catalytic functions on the activation and inhibition of many enzymes, and regulatory functions on cell proliferation, cell cycle, and differentiation. In this study, the effects of magnesium sulfate (MgSO4) on CYP induced ovarian damage were investigated. Immature Wistar-Albino female rats of 28-days were treated with pregnant mare serum gonadotrophin (PMSG) to develop the first generation of preovulatory follicles. Rats of the experimental groups were then treated with either CYP (100 mg/kg, i.p) and MgSO4 (270 mg/kg loading dose; 27 mg/kg maintenance doseX12, i.p) solely or in combination. Following in-vivo 5-bromo-2-deoxyuridine (BrdU) labeling, animals were sacrificed and ovaries were embedded in paraffin and Epon. In the ovaries, added to the evaluation of general morphology and follicle count; BrdU and TUNEL-labeling, cleaved caspase-3 and p27 (cyclin-dependent kinase inhibitor) staining was also performed immunohistochemically and an ultrastructural evaluation was performed by transmission electron microscopy (TEM). The number of primordial follicles were decreased and multilaminar primary and atretic follicles were increased in CYP group. After MgSO4 treatment, while primordial follicle pool were elevated, the number of atretic follicles were decreased. Additionally, decreased BrdU-labeling, increased cleaved caspase 3 immunoreactivity and increased TUNEL labeling were observed in CYP group. In CYP treated animals, observations showed that while MgSO4 administration caused no alterations in BrdU proliferation index and caspase-3 immunoreactivity, it significantly reduced the TUNEL labeling. It was also observed that, while p27 immunoreactivity significantly increased in the nuclei of granulosa and theca cells in the CYP group; MgSO4 treatment significantly reduced these immunoreactivities. The ultrastructural observations showed frequent apoptotic profiles in granulosa and theca cells in both early and advanced stages of follicles in the CYP group and the MgSO4 treatment before the CYP application led to ultrastructural alleviation of the apoptotic process. In conclusion, our data suggest that MgSO4 may provide an option of pharmacologic treatment for fertility preservation owing to the beneficial effects of on chemotherapy-induced accelerated follicular apoptotic process, and the protection of the primordial follicle pool.
Collapse
Affiliation(s)
- Tuğba Ekiz Yılmaz
- Istanbul University, Istanbul Faculty of Medicine, Department of Histology and Embryology, Istanbul 34093, Turkey.
| | - Müge Taşdemir
- Istanbul University, Istanbul Faculty of Medicine, Department of Histology and Embryology, Istanbul 34093, Turkey.
| | - Mehmet Kaya
- Istanbul University, Istanbul Faculty of Medicine, Department of Physiology, Istanbul 34093, Turkey.
| | - Nadir Arıcan
- Istanbul University, Istanbul Faculty of Medicine, Department of Forensic Medicine, Istanbul 34093, Turkey.
| | - Bülent Ahıshalı
- Istanbul University, Istanbul Faculty of Medicine, Department of Histology and Embryology, Istanbul 34093, Turkey.
| |
Collapse
|
42
|
Luo Q, Liu R, Wang L, Hou Y, Zhang H. The Effects of Inhibin B in the Chemotherapy Drug-Induced Premature Ovarian Insufficiency Mice and hPMSCs Treatment. Reprod Sci 2020; 27:1148-1155. [PMID: 31993999 DOI: 10.1007/s43032-019-00128-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 09/25/2019] [Indexed: 12/18/2022]
Abstract
Human placenta mesenchymal stem cells (hPMSCs), have been extensively investigated on the treatment of many diseases. This study was to explore the effects of hPMSCs treatment on the chemotherapy drug-induced premature ovarian insufficiency (POI) mice. Cyclophosphamide (120 mg/kg) and busulfan (30 mg/kg) or cyclophosphamide (70 mg/kg) induced POI mice were used and hPMSCs were injected through tail vein. The hormone levels of serum, morphological changes, the expression and quantitative analysis of inhibin B (INHBB) and FSHR protein, and apoptosis of granulosa cells in ovary were detected. The granulosa cells (GCs) were detected from ovaries of mice and the different concentration of cyclophosphamide on GCs were detected by MTT assay. The apoptosis of GCs was detected by FITC Annexin V Apoptosis Detection Kit. The significant increase in FSH and decrease in E2 and INHBB were observed. Expression of human nuclei was observed in the stroma of ovaries. INHBB and FSHR levels of ovaries were reduced in the POI mice. Following hPMSCs treatment, the amounts of INHBB and FSHR significantly increased close to normal levels. The granulosa cells apoptosis increased in the POI ovaries but decreased after hPMSCs treatment. Moreover, cyclophosphamide has no effect on the GCs and no statistic difference was measured in vitro. The effects of hPMSCs treatment reduce the apoptosis of granulosa cells and restore the ovarian reserve capacity in chemotherapeutic drug-induced POI mice. The data help to further explore new potential clinical therapeutic approach for POI patients.
Collapse
Affiliation(s)
- Qianqian Luo
- School of Basic Medical Sciences&Institute of Reproductive Diseases, Binzhou Medical University, Yantai, Shandong, China
| | - Ranran Liu
- Department of Reproductive Medicine, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Li Wang
- Obstetrical [Maternity] Department, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Yun Hou
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong, China.
| | - Hongqin Zhang
- School of Basic Medical Sciences&Institute of Reproductive Diseases, Binzhou Medical University, Yantai, Shandong, China.
| |
Collapse
|
43
|
Figueroa Gonzalez D, Young F. Gamma Tocopherol Reduced Chemotherapeutic-Induced ROS in an Ovarian Granulosa Cell Line, But Not in Breast Cancer Cell Lines In Vitro. Antioxidants (Basel) 2020; 9:antiox9010051. [PMID: 31936058 PMCID: PMC7023168 DOI: 10.3390/antiox9010051] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/30/2019] [Accepted: 12/31/2019] [Indexed: 01/13/2023] Open
Abstract
Doxorubicin and cyclophosphamide are used to treat breast cancer, but they also cause infertility through off-target cytotoxicity towards proliferating granulosa cells that surround eggs. Each chemotherapeutic generates reactive oxygen species (ROS) but the effects of the combination, or the antioxidants alpha (αToc) and gamma tocopherol (γToc) on ROS in breast cancer or ovarian cells are unknown. Human breast cancer (MCF7, T47D) and ovarian cancer (OVCAR, COV434) cells were loaded with DCDFA and exposed (1, 2, 3, 24 h) to the MCF7-derived EC25 values of individual agents, or to combinations of these. ROS were quantified and viable cells enumerated using crystal violet or DAPI. Each chemotherapeutic killed ~25% of MCF7, T47D and OVCAR cells, but 57 ± 2% (doxorubicin) and 66 ± 2% (cyclophosphamide) of the COV434 granulosa cells. The combined chemotherapeutics decreased COV434 cell viability to 34 ± 5% of control whereas doxorubicin + cyclophosphamide + γToc reduced ROS within 3 h (p < 0.01) and reduced cytotoxicity to 54 ± 4% (p < 0.05). αToc was not cytotoxic, whereas γToc killed ~25% of the breast cancer but none of the ovarian cells. Adding γToc to the combined chemotherapeutics did not change ROS or cytotoxicity in MCF7, T47D or OVCAR cells. The protection γToc afforded COV434 granulosa cells against chemotherapy-induced ROS and cytotoxicity suggests potential for fertility preservation.
Collapse
|
44
|
Ovarian Follicles Rescued 3 Days after Cyclophosphamide Treatment in Adolescent Mice: An Experimental Study Aiming at Maximizing Methods for Fertility Preservation through In Vitro Follicle Culture. Int J Mol Sci 2019; 20:ijms20246190. [PMID: 31817942 PMCID: PMC6940762 DOI: 10.3390/ijms20246190] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 01/30/2023] Open
Abstract
There is currently a lack of knowledge about the feasibility of performing procedures for fertility preservation after chemotherapy treatment has been initiated. In this experimental controlled study using adolescent mice, we aimed to investigate if the chance of rescuing and growing in vitro secondary follicles (SeF) would be affected three days after a single injection of cyclophosphamide (CPA). The main outcomes included were: (1) The number of SeF with good morphologic quality obtained per ovary 3 days after CPA injection, (2) SeF development in culture, (3) small follicle density (SFD) on histology, and (4) apoptosis markers, including terminal deoxynucleotidyl transferase dUTP nick end-labelling (TUNEL), mRNA expression, and distribution of p 53 upregulated modulator of apoptosis (Puma) and phosphatase and tensin homolog (Pten). We found a 60% reduction of SeF obtained per ovary in all CPA-treated groups vs. controls. However, in vitro survival rates at culture day 12 and antrum formation were similar among all groups. On histology, SFD was only significantly reduced in the high CPA dose group. Apoptotic cells were mainly found in large growing follicles of CPA groups. Our study indicates the feasibility of SeF isolation and in vitro follicle culture 3 days following CPA treatment and a still preserved SFD, particularly following a low-dose CPA treatment.
Collapse
|
45
|
Athira VR, Saranya MK, Shivanandappa T, Yajurvedi HN. Multiple dose treatment reduces cyclophosphamide-induced ovarian follicular loss in mice. Birth Defects Res 2019; 112:71-80. [PMID: 31643145 DOI: 10.1002/bdr2.1603] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/18/2019] [Accepted: 09/21/2019] [Indexed: 12/16/2022]
Abstract
This study compares the effects of single dose and multiple dose treatment of cyclophosphamide (CP) on oxidative stress-mediated follicular damage in mouse ovary. In the first experiment, adult female mice were administered with a single dose of CP (100 mg/kg body weight/mouse) and autopsied 72 hr after treatment. In the second experiment, adult female mice were injected with multiple doses of CP (40 mg/kg body weight/day/mouse for 10 consecutive days) and sacrificed on Day 11. There was a 58, 48, 53, and 51% loss of primordial, primary, preantral, and antral follicles, respectively, following the administration of a single dose of CP, whereas, multiple dose of CP caused only 35% reduction in primordial follicles coupled with 28, 23, and 38%, loss of primary, preantral, and antral follicles, respectively. There was a decrease in activities of the ovarian antioxidant enzymes and increase in reactive oxygen species (ROS) and malondialdehyde (MDA) concentrations following single dose CP, whereas multiple dose treatment caused an increase in activities of these enzymes and decrease in ROS and MDA concentrations. The serum concentration of estradiol was significantly decreased following single or multiple dose treatment. The ovarian damage caused by a single high dose of CP administration is higher than that by multiple doses of smaller amount, though the total amount of CP administered was higher with multiple treatment. The results of the current study reveal the benefit of metronomic chemotherapy in cancer treatment, for its effectiveness in reducing ovarian toxicity, a major side effect in young female patients.
Collapse
|
46
|
Roness H, Spector I, Leichtmann-Bardoogo Y, Savino AM, Dereh-Haim S, Meirow D. Pharmacological administration of recombinant human AMH rescues ovarian reserve and preserves fertility in a mouse model of chemotherapy, without interfering with anti-tumoural effects. J Assist Reprod Genet 2019; 36:1793-1803. [PMID: 31250176 PMCID: PMC6730972 DOI: 10.1007/s10815-019-01507-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 06/13/2019] [Indexed: 12/20/2022] Open
Abstract
PURPOSE To determine whether pharmacological administration of recombinant human anti-Mullerian hormone (rAMH) protects the ovarian reserve and preserves fertility without interfering with anti-tumoural cytotoxic action of chemotherapy. METHODS Intraperitoneal delivery of rAMH and ovarian post-receptor activity were assessed with immunohistochemistry and western blot. Differential follicle counts and reproductive outcomes were assessed after cyclophosphamide (Cy) administration, with/without concurrent administration of rAMH. Interference of rAMH with Cy chemotoxicity was assessed on a human breast cancer cell line and an in vivo mouse model of human leukaemia. RESULTS rAMH reached the ovary after intraperitoneal injection and demonstrated post-receptor bioactivity. Cy administration in mice caused primordial follicle activation, as shown by a decrease in primordial follicle population accompanied by an increase in early growing follicles and granulosa cell proliferation. Co-administration of rAMH reduced follicle activation, thereby protecting the primordial follicle reserve, and improving long-term fertility and reproductive outcomes. rAMH co-administration did not interfere with the cytotoxic actions of Cy in vitro on breast cancer cell line or in vivo in a model of human leukaemia. CONCLUSION This study demonstrates that rAMH is bioactive in the ovary for a limited time, and that pharmacological administration of rAMH during chemotherapy treatment reduces follicle activation and primordial follicle loss and significantly improves reproductive outcomes in a mouse model, and does not interfere with the therapeutic actions of the treatment. Further investigation is necessary to determine whether it has similar protective effects in the human ovary.
Collapse
Affiliation(s)
- H Roness
- Fertility Preservation Research Laboratory, Department of Obstetrics and Gynecology, Sheba Medical Centre, Ramat Gan, Israel
| | - I Spector
- Fertility Preservation Research Laboratory, Department of Obstetrics and Gynecology, Sheba Medical Centre, Ramat Gan, Israel
| | - Y Leichtmann-Bardoogo
- Fertility Preservation Research Laboratory, Department of Obstetrics and Gynecology, Sheba Medical Centre, Ramat Gan, Israel
| | - A M Savino
- Department of Human Molecular Genetics and Biochemistry, Sheba Medical Centre, Ramat Gan, Israel
| | - Sanaz Dereh-Haim
- Fertility Preservation Research Laboratory, Department of Obstetrics and Gynecology, Sheba Medical Centre, Ramat Gan, Israel
| | - Dror Meirow
- Fertility Preservation Research Laboratory, Department of Obstetrics and Gynecology, Sheba Medical Centre, Ramat Gan, Israel.
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
47
|
Fertility preservation in patients with hematologic malignancies and recipients of hematopoietic cell transplants. Blood 2019; 134:746-760. [PMID: 31292116 DOI: 10.1182/blood.2018846790] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 05/30/2019] [Indexed: 02/07/2023] Open
Abstract
Patients with hematologic malignancies and those undergoing hematopoietic cell transplantation (HCT) face a complex set of challenges when considering options for fertility preservation (FP). There are no standard options for prepubertal children, and women with hematologic malignancies may not be eligible for standard FP options. Fortunately, initial therapies for most blood cancers are not highly gonadotoxic, affording an important opportunity for postremission counseling and referrals to fertility specialists. These patients face a high risk of relapse, and many will be referred for autologous or allogeneic HCT, which carries an extremely high risk of infertility. The expanding indications for HCT to include benign hematologic disorders as well as autoimmune diseases mandate that all hematologists are familiar with these risks. Oncofertility researchers are continually pushing the boundaries of what may be possible for our patients; in the meantime, communication and shared decision-making between hematologists and patients, as well as program-building, education, and outreach are essential to ensure that these patients, many of whom will be cured, maintain all of their options for a fulfilling life after intensive therapy.
Collapse
|
48
|
Buigues A, Marchante M, Herraiz S, Pellicer A. Diminished Ovarian Reserve Chemotherapy-Induced Mouse Model: A Tool for the Preclinical Assessment of New Therapies for Ovarian Damage. Reprod Sci 2019:1933719119831784. [PMID: 30791852 DOI: 10.1177/1933719119831784] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Diminished ovarian reserve (DOR) and primary ovarian insufficiency (POI) are primary factors leading to infertility. However, there is a lack of appropriate animal models of DOR usable for assessing new therapeutic strategies. In this study, we aimed to evaluate whether chemotherapy treatment in mice could reproduce features similar of that observed in women with DOR. Twenty-one Nonobese diabetic/severe combined immunodeficiency (NOD/SCID) female mice were allocated to 3 groups (n = 7/group): control, single dose of vehicle (Dimethyl Sulfoxide [DMSO]); DOR, single reduced chemotherapy dose; and POI, single standard chemotherapy dose. After 21 days, mice underwent ovarian hyperstimulation and mating. Part of the animals were harvested to analyze ovarian reserve, ovulation and fertilization rates, and morphology, apoptosis, and vascularization of the ovarian stroma. The remaining mice underwent multiple matings to assess pregnancy rates and litter sizes. The DOR and POI mice showed an impaired estrous cyclicity and a decrease in ovarian mass, number of follicles, Metaphase II (MII) oocytes, and embryos as well as in ovarian stroma vascularization. Mice in both models showed also an increase in the percentage of morphologically abnormal follicles, stromal degeneration, and apoptosis. Similar to that observed in DOR and POI patients, these impairments were less severe in DOR than in POI mice. None of the POI females were able to achieve a pregnancy. Meanwhile, DOR females achieved several consecutive pregnancies, although litter size was decreased when compared to controls. In conclusion, a mouse model which displayed most of the ovarian characteristics and fertility outcomes of women with DOR has been established using a single dose of chemotherapy.
Collapse
Affiliation(s)
- Anna Buigues
- 1 IVI Foundation, Valencia Spain
- 2 Department of Pediatrics, Obstetrics and Gynecology, School of Medicine, Valencia University, Valencia, Spain
| | - Maria Marchante
- 1 IVI Foundation, Valencia Spain
- 2 Department of Pediatrics, Obstetrics and Gynecology, School of Medicine, Valencia University, Valencia, Spain
| | - Sonia Herraiz
- 1 IVI Foundation, Valencia Spain
- 3 Reproductive Medicine Research Group, IIS La Fe, Valencia, Spain
- 4 IVI-RMA Valencia, Valencia, Spain
| | - Antonio Pellicer
- 1 IVI Foundation, Valencia Spain
- 3 Reproductive Medicine Research Group, IIS La Fe, Valencia, Spain
- 5 IVI-RMA Rome, Rome, Italy
| |
Collapse
|
49
|
Effect of Chemotherapeutics and Tocopherols on MCF-7 Breast Adenocarcinoma and KGN Ovarian Carcinoma Cell Lines In Vitro. BIOMED RESEARCH INTERNATIONAL 2019; 2019:6146972. [PMID: 30766885 PMCID: PMC6350544 DOI: 10.1155/2019/6146972] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 11/28/2018] [Accepted: 12/30/2018] [Indexed: 02/04/2023]
Abstract
The combination of doxorubicin and cyclophosphamide commonly used to treat breast cancer can cause premature ovarian failure and infertility. α-Tocopherol is a potent antioxidant whereas γ-tocopherol causes apoptosis in a variety of cancer models in vitro including breast cancer. We hypothesised that the combination of doxorubicin (Dox) and 4-hydroperoxycyclophosphamide (4-Cyc) would be more cytotoxic in vitro than each agent alone, and that α-tocopherol would reduce and γ-tocopherol would augment the cytotoxicity of the combined chemotherapeutics. Human MCF-7 breast cancer and KGN ovarian cells were exposed to Dox, 4-Cyc, combined Dox and 4-Cyc, α-tocopherol, γ-tocopherol, or a combination of Dox and 4-Cyc with α-tocopherol or γ–tocopherol. Cell viability was assessed using a crystal violet assay according to four schedules: 24h exposure, 24h exposure + 24h culture in medium, 24h exposure + 48h culture in medium, or 72h continuous exposure. Supernatants from each separate KGN culture experiment (n=3) were examined using an estradiol ELISA. Dox was cytotoxic to both MCF-7 and KGN cells, but 4-Cyc only killed MCF-7 cells. γ-Tocopherol significantly decreased MCF-7 but not KGN cell viability. The combined chemotherapeutics and γ-tocopherol were more cytotoxic to MCF-7 than KGN cells, and α-tocopherol reduced the cytotoxicity of the combined chemotherapeutics towards KGN ovarian cells, but not MCF-7 cells. The addition of both γ-tocopherol and α-tocopherol to the chemotherapeutic combination of Dox and cyclophosphamide has the potential to increase in vitro chemotherapeutic efficacy against breast cancer cells whilst decreasing cytotoxicity towards ovarian granulosa cells.
Collapse
|
50
|
Sonigo C, Beau I, Grynberg M, Binart N. AMH prevents primordial ovarian follicle loss and fertility alteration in cyclophosphamide-treated mice. FASEB J 2018; 33:1278-1287. [PMID: 30113879 DOI: 10.1096/fj.201801089r] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The follicular ovarian reserve, constituted by primordial follicles (PMFs), is established early in life, then keeps declining regularly along reproductive life. The maintenance of a normal female reproductive function implies the presence of a vast amount of dormant PMFs. This process involves a continuous repression of PMF activation into early growing follicle through the balance between factors activating the initiation of follicular growth, mainly actors of the PI3K signaling pathway, and inhibiting factors such as anti-Müllerian hormone (AMH). Any disruption of this balance may induce follicle depletion and subsequent infertility. It has been recently proposed that cyclophosphamide (Cy), an alkylating agent commonly used for treating breast cancer, triggers PMF activation, further leading to premature ovarian insufficiency. Preventing chemotherapy-induced ovarian dysfunction might represent an interesting option for preserving optimal chances of natural or medically assisted conceptions after healing. The aim of the present study was to evaluate, in a model of Cy-treated pubertal mice, whether AMH administration might restrain PMF depletion. The counting of the total PMF number within mouse ovaries showed that recombinant AMH prevented Cy-induced PMF loss. Western blot analysis revealed activation of PI3K signaling pathway after Cy administration. After AMH injection, FOXO3A phosphorylation, a main actor of PMF activation, was significantly decreased. Taken together, these results support a protective role of AMH against Cy-induced follicular loss. We also provide evidence for a possible role of autophagy in the preservation of follicular pool reserve. Therefore, concomitant recombinant AMH administration during chemotherapy might offer a new option for preserving young patients' fertility.-Sonigo, C., Beau, I., Grynberg, M., Binart, N. AMH prevents primordial ovarian follicle loss and fertility alteration in cyclophosphamide-treated mice.
Collapse
Affiliation(s)
- Charlotte Sonigo
- INSERM Unité 1185, Faculté de Médecine Paris-Sud, Université Paris Saclay, Le Kremlin Bicêtre, France; and
| | - Isabelle Beau
- INSERM Unité 1185, Faculté de Médecine Paris-Sud, Université Paris Saclay, Le Kremlin Bicêtre, France; and
| | - Michael Grynberg
- Department of Reproductive Medicine and Fertility Preservation, Hôpital Antoine Béclère, Clamart, France
| | - Nadine Binart
- INSERM Unité 1185, Faculté de Médecine Paris-Sud, Université Paris Saclay, Le Kremlin Bicêtre, France; and
| |
Collapse
|