1
|
Jiang W, Fan S, Zhu Z, Huang H, Tan Y, Peng Y. Design, synthesis and mechanistic studies of novel arylformylhydrazone butylphenyltin complexes as potential anticancer agents. Bioorg Chem 2024; 149:107502. [PMID: 38805912 DOI: 10.1016/j.bioorg.2024.107502] [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: 05/17/2024] [Accepted: 05/25/2024] [Indexed: 05/30/2024]
Abstract
Many diorganotin complexes with various alkyl groups exhibit excellent in vitro anticancer activity. However, most diorganotin is the same alkyl group, and the asymmetric alkyl R group has been rarely reported. Hence, in this paper, twenty butylphenyl mixed dialkyltin arylformylhydrazone complexes have been synthesized by microwave "one-pot" reaction with arylformylhydrazine, substituted α-keto acid or its sodium salt and butylphenyltin dichloride. The crystal structures of nine complexes were determined, indicating that the complexes C1, C2, C11, C12, and C16 ∼ C19 possessed a central symmetric structure of a dinuclear Sn2O2 tetrahedral ring; while the complex C9 is a trinuclear tin-oxygen cluster with a 6-membered ring encased in a 12-membered macrocyclic structure. The inhibiting activity of complexes was tested against the human cell lines NCI-H460, MCF-7, HepG2, Huh-7 and HL-7702. Complex C2 demonstrated the optimal inhibitory effect on HepG2 cells, with an IC50 value of 0.82 ± 0.03 μM. Cellular biology experiments revealed that complex C2 could induce apoptosis and G2/M phase cell cycle arrest in HepG2 and Huh-7 cells. The complex also caused the collapse of the mitochondrial membrane potential and increased intracellular reactive oxygen species in HepG2 and Huh-7 cells. Western blot analysis further clarified that complex C2 could induce cell apoptosis through the mitochondrial pathway along with the release of reactive oxygen species.
Collapse
Affiliation(s)
- Wujiu Jiang
- Key Laboratory of Green Chemistry, Jiangxi Province, College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, Jiangxi 330022, China; Key Laboratory of Functional Metal-Organic Compounds of Hunan Province, Key Laboratory of Organometallic New Materials, College of Hunan Province, College of Chemistry and Materials Science, Hengyang Normal University, Hengyang, Hunan 421008, China.
| | - Shanji Fan
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421000, China
| | - Zhihua Zhu
- Key Laboratory of Functional Metal-Organic Compounds of Hunan Province, Key Laboratory of Organometallic New Materials, College of Hunan Province, College of Chemistry and Materials Science, Hengyang Normal University, Hengyang, Hunan 421008, China
| | - Huifen Huang
- Key Laboratory of Functional Metal-Organic Compounds of Hunan Province, Key Laboratory of Organometallic New Materials, College of Hunan Province, College of Chemistry and Materials Science, Hengyang Normal University, Hengyang, Hunan 421008, China
| | - Yuxing Tan
- Key Laboratory of Functional Metal-Organic Compounds of Hunan Province, Key Laboratory of Organometallic New Materials, College of Hunan Province, College of Chemistry and Materials Science, Hengyang Normal University, Hengyang, Hunan 421008, China
| | - Yiyuan Peng
- Key Laboratory of Green Chemistry, Jiangxi Province, College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, Jiangxi 330022, China.
| |
Collapse
|
2
|
Sriram S, Macedo T, Mavinkurve‐Groothuis A, van de Wetering M, Looijenga LHJ. Alkylating agents-induced gonadotoxicity in prepubertal males: Insights on the clinical and preclinical front. Clin Transl Sci 2024; 17:e13866. [PMID: 38965809 PMCID: PMC11224131 DOI: 10.1111/cts.13866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/26/2024] [Accepted: 06/06/2024] [Indexed: 07/06/2024] Open
Abstract
Rising cure rates in pediatric cancer patients warrants an increased attention toward the long-term consequences of the diagnosis and treatment in survivors. Chemotherapeutic agents can be gonadotoxic, rendering them at risk for infertility post-survival. While semen cryopreservation is an option that can be provided for most (post)pubertal boys before treatment, this is unfortunately not an option prepubertal in age, simply due to the lack of spermatogenesis. Over the last couple of years, studies have thus focused on better understanding the testis niche in response to various chemotherapeutic agents that are commonly administered and their direct and indirect impact on the germ cell populations. These are generally compounds that have a high risk of infertility and have been classified into risk categories in curated fertility guidelines. However, with it comes the lack of evidence and the challenge of using informative models and conditions most reflective of the physiological scenario, in short, the appropriate study designs for clinically relevant outcomes. Besides, the exact mechanism(s) of action for many of these "risk" compounds as well as other agents is unclear. Understanding their behavior and effect on the testis niche will pave the way for incorporating new strategies to ultimately combat infertility. Of the various drug classes, alkylating agents pose the highest risk of gonadotoxicity as per previously established studies as well as risk stratification guidelines. Therefore, this review will summarize the findings in the field of male fertility concerning gonadotoxicity of akylating agents as a result of chemotherapy exposure.
Collapse
Affiliation(s)
- Sruthi Sriram
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | - Tiago Macedo
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | | | | | | |
Collapse
|
3
|
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
|
4
|
Jin Z, Zhao-Xia L, Fan-Ke P, Wen-Juan Z, Min-Li W, Han-Yi Z. Progress in the study of reproductive toxicity of platinum-based antitumor drugs and their means of prevention. Front Pharmacol 2024; 15:1327502. [PMID: 38414732 PMCID: PMC10896984 DOI: 10.3389/fphar.2024.1327502] [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: 10/31/2023] [Accepted: 01/31/2024] [Indexed: 02/29/2024] Open
Abstract
Platinum-based antitumor drugs are broad-spectrum agents with unique mechanisms of action. Combination chemotherapy regimens based on platinum drugs are commonly used in cancer treatment. However, these drugs can cause various adverse reactions in the human body through different routes of administration, including reproductive toxicity, genetic toxicity, and embryonic developmental toxicity. Preventing adverse effects is crucial to enhance patients' quality of life and reduce healthcare costs. This article discusses the types and developmental history of antitumor active platinum compounds, their mechanisms of action, routes of administration, and their potential reproductive, genetic, and embryonic developmental toxicity. This text explores preventive measures based on animal experimental results. Its aim is to provide references for personalized treatment and occupational protection when using platinum drugs. The continuous progress of science and technology, along with the deepening of medical research, suggests that the application of platinum drugs will broaden. Therefore, the development of new platinum drugs will be an important direction for future research.
Collapse
Affiliation(s)
- Zhan Jin
- Gannan Medical University, Ganzhou, China
| | - Liu Zhao-Xia
- Department of Reproductive Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | | | | | - Wei Min-Li
- Department of Reproductive Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Zeng Han-Yi
- Department of Reproductive Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Department of Genetics at the School of Basic Medicine, Gannan Medical University, Ganzhou, China
| |
Collapse
|
5
|
Rosario R, Stewart HL, Spears N, Telfer EE, Anderson RA. Anti-Mullerian hormone attenuates both cyclophosphamide-induced damage and PI3K signalling activation, while rapamycin attenuates only PI3K signalling activation, in human ovarian cortex in vitro. Hum Reprod 2024; 39:382-392. [PMID: 38070496 PMCID: PMC10833070 DOI: 10.1093/humrep/dead255] [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: 05/05/2023] [Revised: 11/13/2023] [Indexed: 02/02/2024] Open
Abstract
STUDY QUESTION What are the effects of cyclophosphamide exposure on the human ovary and can anti-Mullerian hormone (AMH) and rapamycin protect against these? SUMMARY ANSWER Exposure to cyclophosphamide compromises the health of primordial and transitional follicles in the human ovarian cortex and upregulates PI3K signalling, indicating both direct damage and increased follicular activation; AMH attenuates both of these chemotherapy-induced effects, while rapamycin attenuates only PI3K signalling upregulation. WHAT IS KNOWN ALREADY Studies primarily in rodents demonstrate that cyclophosphamide causes direct damage to primordial follicles or that the primordial follicle pool is depleted primarily through excessive initiation of follicle growth. This increased follicular activation is mediated via upregulated PI3K signalling and/or reduced local levels of AMH production due to lost growing follicles. Furthermore, while rodent data show promise regarding the potential benefits of inhibitors/protectants alongside chemotherapy treatment to preserve female fertility, there is no information about the potential for this in humans. STUDY DESIGN, SIZE, DURATION Fresh ovarian cortical biopsies were obtained from 17 healthy women aged 21-41 years (mean ± SD: 31.8 ± 4.9 years) at elective caesarean section. Biopsies were cut into small fragments and cultured for 24 h with either vehicle alone (DMSO), the active cyclophosphamide metabolite 4-hydroperoxycyclophosphamide (4-HC) alone, 4-HC + rapamycin or 4-HC+AMH. Two doses of 4-HC were investigated, 0.2 and 2 μM in separate experiments, using biopsies from seven women (aged 27-41) and six women (aged 21-34), respectively. Biopsies from four women (aged 28-38) were used to investigate the effect of rapamycin or AMH only. PARTICIPANTS/MATERIALS, SETTING, METHODS Histological analysis of ovarian tissue was undertaken for follicle staging and health assessment. Western blotting and immunostaining were used to assess activation of PI3K signalling by measuring phosphorylation of AKT and phosphorylated FOXO3A staining intensity, respectively. MAIN RESULTS AND THE ROLE OF CHANCE Exposure to either dose of 4-HC caused an increase in the proportion of unhealthy primordial (P < 0.0001, both doses) and transitional follicles (P < 0.01 for low dose and P < 0.01 for high dose) compared to vehicle. AMH significantly reduced follicle damage by approximately half in both of the investigated doses of 4-HC (P < 0.0001), while rapamycin had no protective effect on the health of the follicles. Culture with AMH or rapamycin alone had no effect on follicle health. Activation of PI3K signalling following 4-HC exposure was demonstrated by both Western blotting data showing that 4-HC increased in AKT phosphorylation and immunostaining showing increased phosphorylated FOXO3A staining of non-growing oocytes. Treatment with rapamycin reduced the activation of PI3K signalling in experiments with low doses of 4-HC while culture with AMH reduced PI3K activation (both AKT phosphorylation and phosphorylated FOXO3A staining intensity) across both doses investigated. LIMITATIONS, REASONS FOR CAUTION These in vitro studies may not replicate in vivo exposures. Furthermore, longer experiment durations are needed to determine whether the effects observed translate into irreparable deficits of ovarian follicles. WIDER IMPLICATIONS OF THE FINDINGS These data provide a solid foundation on which to explore the efficacy of AMH in protecting non-growing ovarian follicles from gonadotoxic chemotherapies. Future work will require consideration of the sustained effects of chemotherapy treatment and potential protectants to ensure these agents do not impair the developmental competence of oocytes or lead to the survival of oocytes with accumulated DNA damage, which could have adverse consequences for potential offspring. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by grants from TENOVUS Scotland, the Academy of Medical Sciences (to R.R.), the Medical Research Council (G1100357 to R.A.A., MR/N022556/1 to the MRC Centre for Reproductive Health), and Merck Serono UK (to R.A.A.). R.R., H.L.S., N.S., and E.E.T. declare no conflicts of interest. R.A.A. reports grants and personal fees from Roche Diagnostics and Ferring Pharmaceuticals, and personal fees from IBSA and Merck outside the submitted work. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Roseanne Rosario
- Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Hazel L Stewart
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Norah Spears
- Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| | - Evelyn E Telfer
- Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Institute of Cell Biology, University of Edinburgh, Edinburgh, UK
| | - Richard A Anderson
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
6
|
Abd El-Hay RI, Hamed WHE, Mostafa Omar N, Refat El-Bassouny D, Gawish SA. The impact of busulfan on the testicular structure in prepubertal rats: A histological, ultrastructural and immunohistochemical study. Ultrastruct Pathol 2023; 47:424-450. [PMID: 37455400 DOI: 10.1080/01913123.2023.2234470] [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: 05/30/2023] [Accepted: 07/05/2023] [Indexed: 07/18/2023]
Abstract
Busulfan is a widely used cancer chemotherapeutic agent. Temporary or permanent sterility in male patients is one of the most common side effects of this drug. The present study was performed to evaluate the changes in the microscopic structure of the testes of prepubertal rats, as well as the changes in PCNA and caspase-3 immune expression, at different durations after busulfan administration. The rats were 5 weeks old and were divided into two main groups. Control group and busulfan treated group. Busulfan treated group received a single dose of busulfan (40 mg/kg), then animals were subdivided to three subgroups; IIa, IIb, IIc which were sacrificed after four, ten and twenty weeks, respectively, from the beginning of the experiment. Light and electron microscopic studies were done. Serum testosterone level and relative testes weight were assessed. Immunohistochemical staining for anti-proliferating cell nuclear antigen (PCNA) and anti-caspase-3 antigen was also done. Morphometric and statistical studies were carried out. Group II revealed histological and ultrastructural degenerative changes including congested blood vessels and degenerated spermatogenic epithelium, Sertoli cells, and Leydig cells. These changes were more evident after 10 weeks of busulfan administration and were accompanied by absence of mature sperms in the lumen of seminiferous tubules. These changes were associated with a significant reduction in relative testes weight, testosterone level, germinal epithelial height and seminiferous tubule diameter. Moreover, PCNA and caspase-3 immune expression was significantly altered in busulfan treated group. Mild improvement in testicular structure was observed 20 weeks after busulfan treatment.
Collapse
Affiliation(s)
- Reem Ibrahim Abd El-Hay
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Walaa H E Hamed
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Nesreen Mostafa Omar
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Dalia Refat El-Bassouny
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Salwa A Gawish
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
7
|
Almeida GHDR, da Silva-Júnior LN, Gibin MS, Dos Santos H, de Oliveira Horvath-Pereira B, Pinho LBM, Baesso ML, Sato F, Hernandes L, Long CR, Relly L, Miglino MA, Carreira ACO. Perfusion and Ultrasonication Produce a Decellularized Porcine Whole-Ovary Scaffold with a Preserved Microarchitecture. Cells 2023; 12:1864. [PMID: 37508528 PMCID: PMC10378497 DOI: 10.3390/cells12141864] [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: 03/16/2023] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 07/30/2023] Open
Abstract
The application of decellularized scaffolds for artificial tissue reconstruction has been an approach with great therapeutic potential in regenerative medicine. Recently, biomimetic ovarian tissue reconstruction was proposed to reestablish ovarian endocrine functions. Despite many decellularization methods proposed, there is no established protocol for whole ovaries by detergent perfusion that is able to preserve tissue macro and microstructure with higher efficiency. This generated biomaterial may have the potential to be applied for other purposes beyond reproduction and be translated to other areas in the tissue engineering field. Therefore, this study aimed to establish and standardize a protocol for porcine ovaries' decellularization based on detergent perfusion and ultrasonication to obtain functional whole-ovary scaffolds. For that, porcine ovaries (n = 5) were perfused with detergents (0.5% SDS and 1% Triton X-100) and submitted to an ultrasonication bath to produce acellular scaffolds. The decellularization efficiency was evaluated by DAPI staining and total genomic DNA quantification. ECM morphological evaluation was performed by histological, immunohistochemistry, and ultrastructural analyses. ECM physico-chemical composition was evaluated using FTIR and Raman spectroscopy. A cytocompatibility and cell adhesion assay using murine fibroblasts was performed. Results showed that the proposed method was able to remove cellular components efficiently. There was no significant ECM component loss in relation to native tissue, and the scaffolds were cytocompatible and allowed cell attachment. In conclusion, the proposed decellularization protocol produced whole-ovaries scaffolds with preserved ECM composition and great potential for application in tissue engineering.
Collapse
Affiliation(s)
| | | | | | - Henrique Dos Santos
- Department of Physics, State University of Maringá, Maringá 87020-900, Brazil
| | | | - Leticia Beatriz Mazo Pinho
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo 05508-270, Brazil
| | | | - Francielle Sato
- Department of Physics, State University of Maringá, Maringá 87020-900, Brazil
| | - Luzmarina Hernandes
- Department of Morphological Sciences, State University of Maringa, Maringá 87020-900, Brazil
| | - Charles R Long
- Department of Veterinary Physiology and Pharmacology, School of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Luciana Relly
- Department of Veterinary Physiology and Pharmacology, School of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Maria Angelica Miglino
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo 05508-270, Brazil
| | - Ana Claudia Oliveira Carreira
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo 05508-270, Brazil
- Centre for Natural and Human Sciences, Federal University of ABC, Santo André, São Paulo 09210-580, Brazil
| |
Collapse
|
8
|
Matilionyte G, Forsyth G, Guo J, Rimmer MP, Hermann BP, Anderson RA, Mitchell RT. Granulocyte-colony stimulating factor does not prevent in vitro cisplatin-induced germ cell reduction in immature human and mouse testis. BMC Cancer 2023; 23:251. [PMID: 36922758 PMCID: PMC10018904 DOI: 10.1186/s12885-023-10702-y] [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: 12/26/2022] [Accepted: 03/02/2023] [Indexed: 03/18/2023] Open
Abstract
BACKGROUND Currently there are no established fertility preservation options for pre-pubertal boys facing cancer treatment. Granulocyte-colony stimulating factor (G-CSF) treatment has been proposed to be chemoprotective against spermatogonial cell loss in an alkylating chemotherapy model of busulfan treated adult mice. Having previously shown that exposure to the alkylating-like chemotherapy cisplatin resulted in a reduction in germ cell numbers in immature human testicular tissues, we here investigate whether G-CSF would prevent cisplatin-induced germ cell loss in immature human and mouse (fetal and pre-pubertal) testicular tissues. METHODS Organotypic in vitro culture systems were utilised to determine the effects of clinically-relevant concentrations of G-CSF in cisplatin-exposed immature testicular tissues. Human fetal (n = 14 fetuses) and mouse pre-pubertal (n = 4 litters) testicular tissue pieces were cultured and exposed to cisplatin or vehicle control for 24 hrs and analysed at 72 and 240 hrs post-exposure. Combined G-CSF and cisplatin exposure groups explored varying concentrations and duration of G-CSF supplementation to the culture medium (including groups receiving G-CSF before, during and after cisplatin exposure). In addition, effects of G-CSF supplementation alone were investigated. Survival of total germ cell and sub-populations were identified by expression of AP2γ and MAGE-A4 for human gonocytes and (pre)spermatogonia, respectively, and MVH and PLZF, for mouse germ cells and putative spermatogonial stem cells (SSCs) respectively, were quantified. RESULTS Exposure to cisplatin resulted in a reduced germ cell number in human fetal and mouse pre-pubertal testicular tissues at 240 hrs post-exposure. Germ cell number was not preserved by combined exposure with G-CSF using any of the exposure regimens (prior to, during or after cisplatin exposure). Continuous supplementation with G-CSF alone for 14 days did not change the germ cell composition in either human or mouse immature testicular tissues. CONCLUSIONS This study demonstrates that exposure to G-CSF does not prevent cisplatin-induced germ cell loss in immature human and mouse testicular tissues in an in vitro system.
Collapse
Affiliation(s)
- Gabriele Matilionyte
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK
| | - Grace Forsyth
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK
| | - Jingtao Guo
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Michael P Rimmer
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK
| | - Brian P Hermann
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, 1 UTSA Circle, San Antonio, TX, 78249, USA
| | - Richard A Anderson
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK
| | - Rod T Mitchell
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK.
- Royal Hospital for Children & Young People, 50 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK.
| |
Collapse
|
9
|
Almeida GHDR, Iglesia RP, Rinaldi JDC, Murai MK, Calomeno CVAQ, da Silva Junior LN, Horvath-Pereira BDO, Pinho LBM, Miglino MA, Carreira ACO. Current Trends on Bioengineering Approaches for Ovarian Microenvironment Reconstruction. TISSUE ENGINEERING. PART B, REVIEWS 2023. [PMID: 36355603 DOI: 10.1089/ten.teb.2022.0171] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Ovarian tissue has a unique microarchitecture and a complex cellular and molecular dynamics that are essential for follicular survival and development. Due to this great complexity, several factors may lead to ovarian insufficiency, and therefore to systemic metabolic disorders and female infertility. Techniques currently used in the reproductive clinic such as oocyte cryopreservation or even ovarian tissue transplant, although effective, have several limitations, which impair their wide application. In this scenario, mimetic ovarian tissue reconstruction comes as an innovative alternative to develop new methodologies for germ cells preservation and ovarian functions restoration. The ovarian extracellular matrix (ECM) is crucial for oocyte viability maintenance, once it acts actively in folliculogenesis. One of the key components of ovarian bioengineering is biomaterials application that mimics ECM and provides conditions for cell anchorage, proliferation, and differentiation. Therefore, this review aims at describing ovarian tissue engineering approaches and listing the main limitations of current methods for preservation and reestablishment of ovarian fertility. In addition, we describe the main elements that structure this study field, highlighting the main advances and the challenges to overcome to develop innovative methodologies to be applied in reproductive medicine. Impact Statement This review presents the main advances in the application of tissue bioengineering in the ovarian tissue reconstruction to develop innovative solutions for ovarian fertility reestablishment.
Collapse
Affiliation(s)
| | - Rebeca Piatniczka Iglesia
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Mikaelly Kiemy Murai
- Department of Morphological Sciences, State University of Maringa, Maringá, Brazil
| | | | | | | | - Letícia Beatriz Mazo Pinho
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Maria Angelica Miglino
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Ana Claudia Oliveira Carreira
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil.,Center of Natural and Human Sciences, Federal University of ABC, Santo André, Brazil
| |
Collapse
|
10
|
Structural characterization, DNA binding study, antioxidant potential and antitumor activity of diorganotin(IV) complexes against human breast cancer cell line MDA-MB-231. J Organomet Chem 2023. [DOI: 10.1016/j.jorganchem.2023.122671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
|
11
|
Investigation of the female infertility risk associated with anti-cancer therapy. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023:10.1007/s12094-023-03087-8. [PMID: 36689055 DOI: 10.1007/s12094-023-03087-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/11/2023] [Indexed: 01/24/2023]
Abstract
Female infertility is a significant health issue worldwide with a rising incidence. Anti-cancer therapy is one of the most important reasons for increasing infertility. Although anti-cancer treatment increases the rate of survival, it decreases the quality of life through its side effects. The most substantial side effects are sexual dysfunction and infertility. Breast cancer is the most common cancer. The first-line treatment of breast cancer is chemotherapy by alkylating agents like cyclophosphamide, which leads to infertility. For instance, persistent chemotherapy-induced amenorrhea among breast cancer patients could affect almost half of the patients that undergo such therapy. However, some agents or therapeutic methods can ameliorate these intoxicating effects. Chemotherapy plus gonadotropin-releasing hormone agonist, in breast cancer patients, can not only improve overall survival but also reduce ovarian toxicity. Age plays an essential role in chemotherapy-induced amenorrhea. Chemotherapy at a younger age can reduce the risk of infertility. Gynecological cancers including uterine and ovarian cancer, which have high mortality rates, are the most related cancers to infertility. Surgery is the primary treatment of gynecological cancers. Studies demonstrated that fertility-sparing surgery is a better option than radical surgery. In addition, neoadjuvant chemotherapy is mostly a better option than primary cytoreductive surgery in terms of survival and fertility. Immune checkpoint inhibitors (ICIs) have recently played a major role in treating various cancer types. However, ICIs are associated with hypophysitis, which affects ovaries and can lead to infertility. There are some options for ovarian preservation such as embryo cryopreservation, oocyte cryopreservation, ovarian transposition, ovarian tissue cryopreservation, and ovarian suppression by GnRH agonists. Anti-müllerian hormone level can be utilized to monitor the ovarian reserve. Moreover, to avoid fertility loss, approaches such as using transplantation of human placenta mesenchymal stem cells, administrating anti-inflammatory agents and hormone therapy are under investigation.
Collapse
|
12
|
Corpuz-Hilsabeck M, Culty M. Impact of endocrine disrupting chemicals and pharmaceuticals on Sertoli cell development and functions. Front Endocrinol (Lausanne) 2023; 14:1095894. [PMID: 36793282 PMCID: PMC9922725 DOI: 10.3389/fendo.2023.1095894] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/04/2023] [Indexed: 02/01/2023] Open
Abstract
Sertoli cells play essential roles in male reproduction, from supporting fetal testis development to nurturing male germ cells from fetal life to adulthood. Dysregulating Sertoli cell functions can have lifelong adverse effects by jeopardizing early processes such as testis organogenesis, and long-lasting processes such as spermatogenesis. Exposure to endocrine disrupting chemicals (EDCs) is recognized as contributing to the rising incidence of male reproductive disorders and decreasing sperm counts and quality in humans. Some drugs also act as endocrine disruptors by exerting off-target effects on endocrine tissues. However, the mechanisms of toxicity of these compounds on male reproduction at doses compatible with human exposure are still not fully resolved, especially in the case of mixtures, which remain understudied. This review presents first an overview of the mechanisms regulating Sertoli cell development, maintenance, and functions, and then surveys what is known on the impact of EDCs and drugs on immature Sertoli cells, including individual compounds and mixtures, and pinpointing at knowledge gaps. Performing more studies on the impact of mixtures of EDCs and drugs at all ages is crucial to fully understand the adverse outcomes these chemicals may induce on the reproductive system.
Collapse
|
13
|
Gonfloni S, Jodice C, Gustavino B, Valentini E. DNA Damage Stress Response and Follicle Activation: Signaling Routes of Mammalian Ovarian Reserve. Int J Mol Sci 2022; 23:14379. [PMID: 36430860 PMCID: PMC9693393 DOI: 10.3390/ijms232214379] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/14/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Chemotherapy regimens and radiotherapy are common strategies to fight cancer. In women, these therapies may cause side effects such as premature ovarian insufficiency (POI) and infertility. Clinical strategies to protect the ovarian reserve from the lethal effect of cancer therapies needs better understanding of the mechanisms underlying iatrogenic loss of follicle reserve. Recent reports demonstrate a critical role for p53 and CHK2 in the oocyte response to different DNA stressors, which are commonly used to treat cancer. Here we review the molecular mechanisms underlying the DNA damage stress response (DDR) and discuss crosstalk between DDR and signaling pathways implicated in primordial follicle activation.
Collapse
Affiliation(s)
- Stefania Gonfloni
- Department of Biology, University of Rome Tor Vergata, Via Della Ricerca Scientifica, 00133 Rome, Italy
| | - Carla Jodice
- Department of Biology, University of Rome Tor Vergata, Via Della Ricerca Scientifica, 00133 Rome, Italy
| | - Bianca Gustavino
- Department of Biology, University of Rome Tor Vergata, Via Della Ricerca Scientifica, 00133 Rome, Italy
| | - Elvia Valentini
- Department of Biology, University of Rome Tor Vergata, Via Della Ricerca Scientifica, 00133 Rome, Italy
- PhD Program in Cellular and Molecular Biology, 00133 Rome, Italy
| |
Collapse
|
14
|
Zamponi V, La Salvia A, Tarsitano MG, Mikovic N, Rinzivillo M, Panzuto F, Giannetta E, Faggiano A, Mazzilli R. Effect of Neuroendocrine Neoplasm Treatment on Human Reproductive Health and Sexual Function. J Clin Med 2022; 11:jcm11143983. [PMID: 35887747 PMCID: PMC9324753 DOI: 10.3390/jcm11143983] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 02/02/2023] Open
Abstract
Neuroendocrine neoplasms (NEN) are characterized by a wide clinical heterogeneity and biological variability, with slow progression and long survival in most cases. Although these tumors can affect young adults, there are few studies that focus on the sexual and reproductive system. The aim of this review was to evaluate the effect of NEN treatment, including somatostatin analogues (SSA), targeted therapy (Everolimus and Sunitinib), radiolabeled-SSA and chemotherapy, on male and female reproductive systems and sexual function. This narrative review was performed for all available prospective and retrospective studies, case reports and review articles published up to March 2022 in PubMed. To date, few data are available on the impact of SSA on human fertility and most of studies come from acromegalic patients. However, SSAs seem to cross the blood–placental barrier; therefore, pregnancy planning is strongly recommended. Furthermore, the effect of targeted therapy on reproductive function is still undefined. Conversely, chemotherapy has a well-known negative impact on male and female fertility. The effect of temozolomide on reproductive function is still undefined, even if changes in semen parameters after the treatment have been described. Finally, very few data are available on the sexual function of NEN treatment.
Collapse
Affiliation(s)
- Virginia Zamponi
- Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, 00185 Rome, Italy; (V.Z.); (N.M.); (A.F.); (R.M.)
| | - Anna La Salvia
- Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
- Correspondence: ; Tel.: +39-0652665698
| | - Maria Grazia Tarsitano
- Department of Medical and Surgical Science, University Magna Graecia, 88100 Catanzaro, Italy;
| | - Nevena Mikovic
- Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, 00185 Rome, Italy; (V.Z.); (N.M.); (A.F.); (R.M.)
| | - Maria Rinzivillo
- Digestive Disease Unit, ENETS Center of Excellence, Sant’Andrea University Hospital, 00189 Rome, Italy; (M.R.); (F.P.)
| | - Francesco Panzuto
- Digestive Disease Unit, ENETS Center of Excellence, Sant’Andrea University Hospital, 00189 Rome, Italy; (M.R.); (F.P.)
- Department of Medical-Surgical Sciences and Translational Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Elisa Giannetta
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy;
| | - Antongiulio Faggiano
- Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, 00185 Rome, Italy; (V.Z.); (N.M.); (A.F.); (R.M.)
| | - Rossella Mazzilli
- Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, 00185 Rome, Italy; (V.Z.); (N.M.); (A.F.); (R.M.)
| |
Collapse
|
15
|
Delessard M, Stalin L, Rives-Feraille A, Moutard L, Saulnier J, Dumont L, Rives N, Rondanino C. Achievement of complete in vitro spermatogenesis in testicular tissues from prepubertal mice exposed to mono- or polychemotherapy. Sci Rep 2022; 12:7407. [PMID: 35523907 PMCID: PMC9076692 DOI: 10.1038/s41598-022-11286-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/18/2022] [Indexed: 02/08/2023] Open
Abstract
The assessment of the impact of chemotherapies on in vitro spermatogenesis in experimental models is required before considering the application of this fertility restoration strategy to prepubertal boys who received these treatments before testicular tissue cryopreservation. The present work investigated the effects of exposure of prepubertal mice to mono- (vincristine or cyclophosphamide) and polychemotherapy (a combination of vincristine and cyclophosphamide) on the first wave of in vitro spermatogenesis. When testicular tissue exposed to monochemotherapy was preserved, polychemotherapy led to severe alterations of the seminiferous epithelium and increased apoptosis in prepubertal testes prior in vitro maturation, suggesting a potential additive gonadotoxic effect. These alterations were also found in the testicular tissues of polychemotherapy-treated mice after 30 days of organotypic culture and were associated with a reduction in the germ cell/Sertoli cell ratio. The different treatments neither altered the ability of spermatogonia to differentiate in vitro into spermatozoa nor the yield of in vitro spermatogenesis. However, more spermatozoa with morphological abnormalities and fragmented DNA were produced after administration of polychemotherapy. This work therefore shows for the first time the possibility to achieve a complete in vitro spermatogenesis after an in vivo exposure of mice to a mono- or polychemotherapy before meiotic entry.
Collapse
Affiliation(s)
- Marion Delessard
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France
| | - Laura Stalin
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France
| | - Aurélie Rives-Feraille
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France
| | - Laura Moutard
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France
| | - Justine Saulnier
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France
| | - Ludovic Dumont
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France
| | - Nathalie Rives
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France
| | - Christine Rondanino
- INSERM, U1239, Team Adrenal and Gonadal Pathophysiology, Laboratory of Neuroendocrine Endocrine and Germinal Differentiation and Communication, Rouen University Hospital, Rouen Normandy University, 76000, Rouen, France.
| |
Collapse
|
16
|
Levi M, Ben-Aharon I, Shalgi R. Irinotecan (CPT-11) Treatment Induces Mild Gonadotoxicity. FRONTIERS IN REPRODUCTIVE HEALTH 2022; 4:812053. [PMID: 36303648 PMCID: PMC9580821 DOI: 10.3389/frph.2022.812053] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 02/15/2022] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND Gonadal toxicity following chemotherapy is an important issue among the population of young cancer survivors. The inhibitor of DNA topoisomerase I, irinotecan (CPT-11), is widely used for several cancer types. However, little is known about the effect of irinotecan on the fertility of both genders. Thus, the aim of the present study was to evaluate irinotecan gonadotoxicity, using a mouse model. METHODS Mature male and female mice were injected intraperitoneally with either saline (), irinotecan (100 mg/kg) or cyclophosphamide (100 mg/kg); and sacrificed one week or three months later for an acute or long-term toxicity assessment, respectively. We used thorough and advanced fertility assessment by already established methods: Gonadal and epididymal weights, as well as sperm count and sperm motility were determined; serum anti-Müllerian hormone (AMH) was measured by ELISA. Immunohistochemistry (Ki-67), immunofluorescence (PCNA, CD34), terminal transferase-mediated deoxyuridine 5-triphosphate nick-end labeling (TUNEL) and computerized analysis were performed to examine gonadal proliferation, apoptosis and vascularization. qPCR was used to assess the amount of testicular spermatogonia (Id4 and Gafra1 mRNA) and ovarian primordial oocytes reserves (Sohlh2, Nobox and Figla mRNA). RESULTS Females: Irinotecan administration induced acute ovarian apoptosis and decreased vascularity, as well as a mild, statistically significant, long-term decrease in the number of growing follicles, ovarian weight, and ovarian reserve. Males: Irinotecan administration caused an acute testicular apoptosis and reduced testicular spermatogenesis, but had no effect on vascularity. Irinotecan induced long-term decrease of testicular weight, sperm count and testicular spermatogonia and caused elevated serum AMH. CONCLUSION Our findings imply a mild, though irreversible effect of irinotecan on mice gonads.
Collapse
Affiliation(s)
- Mattan Levi
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- IVF Unit, Meir Medical Center, Kfar Saba, Israel
| | - Irit Ben-Aharon
- Division of Oncology, Rambam Health Care Campus, Haifa, Israel
- Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Ruth Shalgi
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
17
|
Ntemou E, Vidal PD, Alexandri C, Van den Steen G, Lambertini M, Demeestere I. Ovarian toxicity of carboplatin and paclitaxel in mouse carriers of mutation in BRIP1 tumor suppressor gene. Sci Rep 2022; 12:1658. [PMID: 35105904 PMCID: PMC8807594 DOI: 10.1038/s41598-022-05357-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 01/11/2022] [Indexed: 11/19/2022] Open
Abstract
More than 10% of women diagnosed with breast cancer during reproductive age carry hereditary germline pathogenic variants in high-penetrance BRCA genes or in others genes involved in DNA repair mechanisms such as PALB2, BRIP or ATM. Anticancer treatments may have an additional negative impact on the ovarian reserve and subsequently on the fertility of young patients carrying such mutations. Recently, the combination of carboplatin and paclitaxel is being recommended to these BRCA-mutated patients as neoadjuvant therapy. However, the impact on the ovary is unknown. Here, we investigated their effect of on the ovarian reserve using mice carriers of BRCA1-interacting protein C-terminal helicase-1 (BRIP1) mutation that plays an important role in BRCA1-dependent DNA repair. Results revealed that the administration of carboplatin or paclitaxel did not affect the ovarian reserve although increased DNA double-strand breaks were observed with carboplatin alone. Co-administration of carboplatin and paclitaxel resulted in a significant reduction of the ovarian reserve leading to a lower IVF performance, and an activation of the PI3K-Pten pathway, irrespective of the genetic background. This study suggests that co-administration of carboplatin and paclitaxel induces cumulative ovarian damage and infertility but a heterozygote genetic predisposition for DNA damage related to BRCA1 gene function does not increase this risk.
Collapse
Affiliation(s)
- E Ntemou
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles, Brussels, Belgium
| | - P Diaz Vidal
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles, Brussels, Belgium
| | - C Alexandri
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles, Brussels, Belgium
| | - G Van den Steen
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles, Brussels, Belgium
| | - M Lambertini
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genoa, Italy
- Department of Medical Oncology, UOC Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - I Demeestere
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles, Brussels, Belgium.
- Obstetrics and Gynaecology Department, Erasme Hospital, Brussels, Belgium.
| |
Collapse
|
18
|
Jiang W, Zhang Z, Ni P, Tan Y. OUP accepted manuscript. Metallomics 2022; 14:6585273. [PMID: 35556135 DOI: 10.1093/mtomcs/mfac021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 03/24/2022] [Indexed: 11/14/2022]
Abstract
Fourteen new organotin(IV) complexes were successfully synthesized and characterized by elemental analyses, Fourier transform infrared spectroscopy (FT-IR), multinuclear (1H, 13C, and 119Sn) NMR spectroscopy, high-resolution mass spectrometry (HRMS), and X-ray single-crystal techniques. Crystallographic data showed that the complexes 1b, 2b, 3b, and 5b were macrocyclic compounds, 4b exhibited a one-dimensional spiral chain structure with distorted trigonal bipyramidal geometry, other complexes were centrosymmetric dimers, and there was an Sn2O2 four-membered ring in the middle of the molecule. In-vitro anticancer activity against the three human tumor cell lines NCI-H460, MCF-7, and HepG2 was studied, and the dibutyltin complex 5a is a more potent antitumor agent than other complexes and cisplatin. Cell apoptosis study of 5a with the highest activity on HepG2 cancer cell lines was done by flow cytometry; it was shown that the antitumor activity of 5a was related to apoptosis, and it inhibited proliferation by blocking cells in the G2/M phase. The single-cell gel electrophoresis assay results show that 5a induces DNA damage. 5a interacts with ct-DNA by intercalating the mode of interaction. UV-visible absorption spectrometry, fluorescence competitive, viscosity measurements, and gel electrophoresis results also support the intercalative mode of interaction for 5a with DNA.
Collapse
Affiliation(s)
- Wujiu Jiang
- Key Laboratory of Functional Metal-Organic Compounds of Hunan Province, Key Laboratory of Organometallic New Materials, College of Hunan Province, Hunan Provincial Engineering Research Center for Monitoring and Treatment of Heavy Metals Pollution in the Upper Reaches of XiangJiang River, College of Chemistry and Materials Science, Hengyang Normal University, Hengyang, Hunan 421008, China
| | - Zhijian Zhang
- Key Laboratory of Functional Metal-Organic Compounds of Hunan Province, Key Laboratory of Organometallic New Materials, College of Hunan Province, Hunan Provincial Engineering Research Center for Monitoring and Treatment of Heavy Metals Pollution in the Upper Reaches of XiangJiang River, College of Chemistry and Materials Science, Hengyang Normal University, Hengyang, Hunan 421008, China
| | - Penghui Ni
- Key Laboratory of Functional Metal-Organic Compounds of Hunan Province, Key Laboratory of Organometallic New Materials, College of Hunan Province, Hunan Provincial Engineering Research Center for Monitoring and Treatment of Heavy Metals Pollution in the Upper Reaches of XiangJiang River, College of Chemistry and Materials Science, Hengyang Normal University, Hengyang, Hunan 421008, China
| | - Yuxing Tan
- Key Laboratory of Functional Metal-Organic Compounds of Hunan Province, Key Laboratory of Organometallic New Materials, College of Hunan Province, Hunan Provincial Engineering Research Center for Monitoring and Treatment of Heavy Metals Pollution in the Upper Reaches of XiangJiang River, College of Chemistry and Materials Science, Hengyang Normal University, Hengyang, Hunan 421008, China
| |
Collapse
|
19
|
Jiang W, Tan Y, Peng Y. Aroylhydrazone Diorganotin Complexes Causes DNA Damage and Apoptotic Cell Death: From Chemical Synthesis to Biochemical Effects. Int J Mol Sci 2021; 22:ijms222413525. [PMID: 34948323 PMCID: PMC8709053 DOI: 10.3390/ijms222413525] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/09/2021] [Accepted: 12/14/2021] [Indexed: 12/25/2022] Open
Abstract
Under microwave irradiation, eighteen new aroylhydrazone diorganotin complexes (1a–9b) were produced through the reaction of aroylhydrazine, 2-ketobutyric acid, and the corresponding diorganotin. Fourier transform infrared spectroscopy, 1H, 13C, and 119Sn nuclear magnetic resonance spectroscopies, high-resolution mass spectroscopy, X-ray crystallography, and thermogravimetric analysis (TGA) were performed to characterize the complexes. The in vitro anticancer activity for complexes were assessed using a CCK-8 assay on human cancer cells of HepG2, NCI-H460, and MCF-7. Complex 4b revealed more intensive anticancer activity against MCF-7 cells than the other complexes and cisplatin. Flow cytometry analysis and transmission electron microscope observation demonstrated that complex 4b mediated cell apoptosis of MCF-7 cells and arrested cell cycle in S phase. Western blotting analysis showed that 4b induced DNA damage in MCF-7 cells and led to apoptosis by the ATM-CHK2-p53 pathway. The single cell gel electrophoreses assay results showed that 4b induced DNA damage. The DNA binding activity of 4b was studied by UV–Visible absorption spectrometry, fluorescence competitive, viscosity measurements, gel electrophoresis, and molecular docking, and the results show that 4b can be well embedded in the groove and cleave DNA.
Collapse
Affiliation(s)
- Wujiu Jiang
- Key Laboratory of Functional Small Organic Molecule, Ministry of Education, Key Laboratory of Green Chemistry, College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang 330022, China;
- Hunan Provincial Engineering Research Center for Monitoring and Treatment of Heavy Metals Pollution in the Upper Reaches of XiangJiang River, Key Laboratory of Functional Metal-Organic Compounds of Hunan Province, Key Laboratory of Functional Organometallic Materials, College of Chemistry and Materials Science, University of Hunan Province, Hengyang 421008, China
- Correspondence: (W.J.); (Y.P.)
| | - Yuxing Tan
- Key Laboratory of Functional Small Organic Molecule, Ministry of Education, Key Laboratory of Green Chemistry, College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang 330022, China;
- Hunan Provincial Engineering Research Center for Monitoring and Treatment of Heavy Metals Pollution in the Upper Reaches of XiangJiang River, Key Laboratory of Functional Metal-Organic Compounds of Hunan Province, Key Laboratory of Functional Organometallic Materials, College of Chemistry and Materials Science, University of Hunan Province, Hengyang 421008, China
| | - Yiyuan Peng
- Key Laboratory of Functional Small Organic Molecule, Ministry of Education, Key Laboratory of Green Chemistry, College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang 330022, China;
- Correspondence: (W.J.); (Y.P.)
| |
Collapse
|
20
|
Poojary KK, Nayak G, Vasani A, Kumari S, Dcunha R, Kunhiraman JP, Gopalan D, Rao RR, Mutalik S, Kalthur SG, Murari MS, Raghu SV, Adiga SK, Kalthur G. Curcumin nanocrystals attenuate cyclophosphamide-induced testicular toxicity in mice. Toxicol Appl Pharmacol 2021; 433:115772. [PMID: 34715073 DOI: 10.1016/j.taap.2021.115772] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 10/09/2021] [Accepted: 10/18/2021] [Indexed: 12/11/2022]
Abstract
The cancer therapy using cyclophosphamide (CP) has been associated with adverse effects on the testicular function that raises concerns about the future fertility potential among cancer survivors. Curcumin, a polyphenol, has shown to possess a plethora of biological functions including tissue protective effects. In the present study, we investigated the protective effects of curcumin nanocrystals (NC) in mitigation of CP-induced testicular toxicity. Healthy adult (8-10 week) and prepubertal (2 week) male Swiss albino mice were injected with a single dose of CP (200 mg/kg) intraperitoneally (i.p). NC (4 mg/kg, i.p.) was administered every alternate day, for 35 days in adult mice while, a single dose of NC was injected intraperitoneally to prepubertal mice 1 h prior to CP. Administration of multiple doses of NC ameliorated CP-induced testicular toxicity in adult mice, which was evident from the improved sperm functional competence, sperm chromatin condensation, seminiferous tubule architecture and decreased apoptosis in testicular cells. Further, administration of NC 1 h prior to CP in prepubertal mice modulated the expression of genes pertaining to proliferation, pluripotency, DNA damage and DNA repair in spermatogonial cells at 24 h after the treatment. Overall, these results suggest that NC could be a promising chemoprotective agent, which can have potential application in male fertility preservation.
Collapse
Affiliation(s)
- Keerthana Karunakar Poojary
- Division of Clinical Embryology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India
| | - Guruprasad Nayak
- Division of Reproductive Biology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India
| | - Ashna Vasani
- Division of Reproductive Biology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India
| | - Sandhya Kumari
- Division of Reproductive Biology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India
| | - Reyon Dcunha
- Division of Reproductive Biology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India
| | - Jyolsna Ponnaratta Kunhiraman
- Division of Reproductive Biology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India
| | - Divya Gopalan
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Rajat Radhakrishna Rao
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Sneha Guruprasad Kalthur
- Department of Anatomy, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India
| | - M S Murari
- DST PURSE Program, Mangalore University, Mangalagangotri 574199, Karnataka, India
| | - Shamprasad Varija Raghu
- Neurogenetics Lab, Department of Applied Zoology, Mangalore University, Mangalagangotri 574199, Karnataka, India
| | - Satish Kumar Adiga
- Division of Clinical Embryology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India
| | - Guruprasad Kalthur
- Division of Reproductive Biology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India.
| |
Collapse
|
21
|
Female Oncofertility: Current Understandings, Therapeutic Approaches, Controversies, and Future Perspectives. J Clin Med 2021; 10:jcm10235690. [PMID: 34884393 PMCID: PMC8658080 DOI: 10.3390/jcm10235690] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 11/25/2021] [Accepted: 12/02/2021] [Indexed: 12/14/2022] Open
Abstract
Recent advances in early detection and oncological therapies have ameliorated the survival rate of young cancer patients. Yet, ovarian impairment induced by chemotherapy and radiotherapy is still a challenging issue. This review, based on clinical and lab-based studies, summarizes the evidence of gonadotoxicity of chemoradiotherapy, the recent approaches, ongoing controversies, and future perspectives of fertility preservation (FP) in female patients who have experienced chemo- or radio-therapy. Existing data indicate that chemotherapeutic agents induce DNA alterations and massive follicle activation via the phosphoinositide 3-kinase (PI3K)/Akt signaling pathway. Meanwhile, the radiation causes ionizing damage, leading to germ cell loss. In addition to the well-established methods, numerous therapeutic approaches have been suggested, including minimizing the follicle loss in cryopreserved ovarian grafts after transplantation, in vitro activation or in vitro growing of follicles, artificial ovarian development, or fertoprotective adjuvant to prevent ovarian damage from chemotherapy. Some reports have revealed positive outcomes from these therapies, whereas others have demonstrated conflictions. Future perspectives are improving the live birth rate of FP, especially in patients with adverse ovarian reserve, eliminating the risk of malignancy reintroducing, and increasing society’s awareness of FP importance.
Collapse
|
22
|
Pairoj S, Damrongsak P, Damrongsak B, Jinawath N, Kaewkhaw R, Ruttanasirawit C, Leelawattananon T, Locharoenrat K. Antitumor activities of carboplatin-doxorubicin-ZnO complexes in different human cancer cell lines (breast, cervix uteri, colon, liver and oral) under UV exposition. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2021; 49:120-135. [PMID: 33491496 DOI: 10.1080/21691401.2021.1876718] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 01/10/2021] [Indexed: 01/01/2023]
Abstract
This study aimed to examine the pharmacological profiles of multiple chemo drug candidates in systematic circulation to enhance their specific interactions with five human cancer cell lines. ZnO nanoparticles were successfully bound with chemo drugs via physical adsorption. The drug loading capacity was confirmed by FTIR, whereas the loading efficiency was determined via UV-vis spectrometry. The mean hydrodynamic size increased to 69-82 nm after chemo-drug immobilization via non-covalent interaction with ZnO. Among the nine formulated chemo drugs, the carboplatin (CP)-doxorubicin (DOX)-ZnO complex under UV light irradiation exhibited high sensitivity towards human breast adenocarcinoma cells without affecting human keratinocyte immortal cells with an IC50 of 0.137 µg/mL, whereas the loading capacity and efficiency of CP-DOX-ZnO were 77.81% and 99.05%, respectively. Fluorescence images confirmed that CP-DOX-ZnO using DOX served as a fluorescence enhancer specifically bound onto the cell membranes, which became almost saturated after 24 h incubation. Carboplatin-DOX-ZnO was possibly endocytosed by cancer cells and was selectively internalized into the target cells; thus, free chemo drug was released in the cytoplasm, which induced acute apoptosis. This resulted in complete inhabitation of growth signal of target cancer cells.
Collapse
Affiliation(s)
- Suttirak Pairoj
- Department of Physics, Faculty of Science, Biomedical Physics Research Unit, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand
| | - Pattareeya Damrongsak
- Department of Physics, Faculty of Science, Biomedical Physics Research Unit, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand
| | - Badin Damrongsak
- Department of Physics, Faculty of Science, Silpakorn University, Nakornpathom, Thailand
| | - Natini Jinawath
- Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Bangkok, Thailand
| | - Rossukon Kaewkhaw
- Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Bangkok, Thailand
| | - Chinnapat Ruttanasirawit
- Department of Physics, Faculty of Science, Biomedical Physics Research Unit, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand
| | - Tanaporn Leelawattananon
- Department of Physics, Faculty of Science, Biomedical Physics Research Unit, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand
| | - Kitsakorn Locharoenrat
- Department of Physics, Faculty of Science, Biomedical Physics Research Unit, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand
| |
Collapse
|
23
|
Zhao H, Gu W, Pan W, Zhang H, Shuai L, Diao R, Wang L. [miR-483-5p aggravates cisplatin-induced premature ovarian insufficiency in rats by targeting FKBP4]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:801-810. [PMID: 34238731 DOI: 10.12122/j.issn.1673-4254.2021.06.01] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
OBJECTIVE To investigate the role of FKBP4 protein in cisplatin-induced premature ovarian insufficiency (POI). OBJECTIVE We performed ITRAQ assay of the ovarian tissues from 4 mice with cisplatin-induced POI and 4 control mice, and identified FKBP4 as a significantly down-regulated protein in the oocytes and granulosa cells following cisplatin treatment. TargetScan software was used for target analysis of FKBP4, and qRT-PCR and Western blotting were used to verify the expression levels of miR-483-5p and FKBP4 in the mouse models. Serum samples were collected from patients with POI and healthy women for detecting miR-483-5p level with qRT-PCR. Cell transfection and dual-luciferase assay were performed to determine the relationship between miR-483-5p and FKBP4. In primary granulosa cells and KGN cells, we examined the effect of miR-483-5p alone, miR-483-5p and cisplatin, and miR-483-5p combined with both cisplatin and FKBP4 on cell apoptosis. We also assessed ovarian function in a transgenic mouse model with ovarian miR-483-5p overexpression in comparison wigh wildtype mice using immunofluorescence assay, in situ hybridization and ELISA. OBJECTIVE Ovarian FKBP4 expression was significantly decreased in mice with cisplatin-induced POI. Analysis using TargetScan software indicated that FKBP4 was the potential target of miR-483-5p, which was highly expressed in the ovaries and serum of POI mice and in the serum of patients with POI. In vitro experiments further confirmed that FKBP4 was the target of miR-483-5p. In KGN and primary granulosa cells, FKBP4 overexpression significantly reduced cell apoptosis induced by both cisplatin and miR-483-5p overexpression (P= 0.0045 and 0.0177, respectively). In the transgenic mice with miR-483-5p overexpression in the oocytes, cisplatin induced more severe ovarian damages as compared with those in the wild-type mice. OBJECTIVE miR-483-5p/FKBP4 is a new and important pathway in cisplatin-induced POI, in which cisplatin increases ovarian miR- 483-5p expression to result in targeted downregulation of FKBP4. Up-regulation of miR-483-5p may increase ovarian sensitivity to cisplatin and cause severe ovarian dysfunction. Detection of serum miR-483-5p level may help to predict the occurrence and development of POI.
Collapse
Affiliation(s)
- H Zhao
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - W Gu
- Department of Biobank, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - W Pan
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - H Zhang
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, China
| | - L Shuai
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - R Diao
- Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - L Wang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.,Shenzhen Second People's Hospital, Shenzhen 518035, China
| |
Collapse
|
24
|
Tian En L, Brougham MFH, Wallace WHB, Mitchell RT. Impacts of platinum-based chemotherapy on subsequent testicular function and fertility in boys with cancer. Hum Reprod Update 2021; 26:874-885. [PMID: 32935838 PMCID: PMC7600277 DOI: 10.1093/humupd/dmaa041] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/10/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Children with cancer often face infertility as a long-term complication of their treatment. For boys, compromised testicular function is common after chemotherapy and currently there are no well-established options to prevent this damage. Platinum-based agents are used to treat a wide variety of childhood cancers. However, platinum agents are not currently included in the cyclophosphamide equivalent dose (CED), which is used clinically to assess the risks to fertility posed by combination chemotherapy in children with cancer. OBJECTIVE AND RATIONALE This was a systematic search of the literature designed to determine the evidence for effects of platinum-based cancer treatment on the prepubertal human testis in relation to subsequent testicular function and fertility. SEARCH METHODS PubMed and EMBASE were searched for articles published in English between 01 January 1966 and 05 April 2020 using search terms including 'cancer treatment', 'chemotherapy', 'human', 'prepubertal', 'testis', 'germ cells', 'testosterone' and related terms. Abstracts were screened and full-text articles were obtained for those that met the three major inclusion criteria (age ≤12 years at treatment, exposure to platinum-based chemotherapeutic and measure of reproductive function). Screening of bibliographies for full-text articles was used to identify additional studies. OUTCOMES Our initial search identified 1449 articles of which 20 (1.3%) studies (n = 13 759 males) met all inclusion criteria. A control group (healthy individuals or siblings) was included for 5/20 (25%) studies. A total of 10/20 (50%) studies provided sub-analysis of the relative gonadotoxicity of platinum-based agents.The primary outcome measures were: pregnancies and fatherhood; semen analysis; and hormonal function. For pregnancies and fatherhood, three studies (n = 10 453 males) reported negative associations with platinum-agents, including the largest (n = 5640) controlled study (hazard ratio = 0.56, P = 0.0023), whilst two other studies (n = 1781) with platinum sub-analysis reported no association. For semen analysis (based on World Health Organization criteria), platinum-based chemotherapy was associated with azoospermia in one study (n = 129), whilst another (n = 44) found no association and the remainder did not perform platinum-based sub-analysis. For hormone analysis, conflicting results were obtained regarding potential associations between platinum-based agents and elevated FSH (a proxy for impaired spermatogenesis); however, the majority of these studies were based on low numbers of patients receiving platinum-based chemotherapy. WIDER IMPLICATIONS Overall, these results indicate that platinum-based chemotherapy should be included in clinical calculators, for example CED, used to determine gonadotoxicity for childhood cancer treatment. These findings have important implications for clinicians regarding counselling patients and their carer(s) on fertility risk, guiding requirements for fertility preservation strategies (e.g. testicular tissue cryopreservation) and modification of treatments to reduce or eliminate the risk of infertility in childhood cancer survivors.
Collapse
Affiliation(s)
- Lim Tian En
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Mark F H Brougham
- Department of Paediatric Oncology, Royal Hospital for Sick Children, Edinburgh, UK
| | | | - Rod T Mitchell
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK.,Department of Paediatric Endocrinology, Royal Hospital for Sick Children, Edinburgh, UK
| |
Collapse
|
25
|
Lopes F, Tholeti P, Adiga SK, Anderson RA, Mitchell RT, Spears N. Chemotherapy induced damage to spermatogonial stem cells in prepubertal mouse in vitro impairs long-term spermatogenesis. Toxicol Rep 2020; 8:114-123. [PMID: 33425685 PMCID: PMC7782321 DOI: 10.1016/j.toxrep.2020.12.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/24/2020] [Accepted: 12/21/2020] [Indexed: 11/27/2022] Open
Abstract
Chemotherapy can affect testis development of young boys with cancer, reducing the chances of fatherhood in adulthood. Studies using experimental models are needed to determine the damage caused by individual chemotherapy drugs in order to predict the risk of infertility and direct patients towards appropriate fertility preservation options. Here, we investigated the individual role of two drugs, cisplatin and doxorubicin, using an in vitro culture model of prepubertal (postnatal day 5) mouse testis that supports induction and maintenance of full spermatogenesis. Twenty-four hour exposure with either drug at clinically-relevant doses (0.25, 0.5 or 0.75 μg/mL for cisplatin, or 0.01, 0.03 or 0.05 μg/mL for doxorubicin), induced an acute significant loss of spermatogonial stem cells (SSCs; PLZF+), proliferating SSCs (PLZF+BrdU+), total germ cells (MVH+), and spermatocytes (SCP3+) one week after chemotherapy exposure. By the time of the first (Week 4) and second (Week 8) waves of spermatogenesis, there was no longer any effect on SSC or proliferating SSC numbers in drug-exposed testis compared to untreated tissue: however, the populations of total germ cells and spermatocytes were still lower in the higher-dose cisplatin treated groups, along with a reduced frequency of round and elongated spermatids in both cisplatin- and doxorubicin-treated testis fragments. Overall, this study details a direct impairment of germ cell development following acute chemotherapy-induced damage during the prepubertal phase, most likely due to an effect on SSCs, using an in vitro culture system that successfully recapitulates key events of mouse spermatogenesis.
Collapse
Affiliation(s)
- Federica Lopes
- Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - Prathima Tholeti
- Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Department of Clinical Embryology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Satish K. Adiga
- Department of Clinical Embryology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | | | - Rod T. Mitchell
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - Norah Spears
- Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
26
|
Wyns C, Kanbar M, Giudice MG, Poels J. Fertility preservation for prepubertal boys: lessons learned from the past and update on remaining challenges towards clinical translation. Hum Reprod Update 2020; 27:433-459. [PMID: 33326572 DOI: 10.1093/humupd/dmaa050] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 09/25/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Childhood cancer incidence and survivorship are both on the rise. However, many lifesaving treatments threaten the prepubertal testis. Cryopreservation of immature testicular tissue (ITT), containing spermatogonial stem cells (SSCs), as a fertility preservation (FP) option for this population is increasingly proposed worldwide. Recent achievements notably the birth of non-human primate (NHP) progeny using sperm developed in frozen-thawed ITT autografts has given proof of principle of the reproductive potential of banked ITT. Outlining the current state of the art on FP for prepubertal boys is crucial as some of the boys who have cryopreserved ITT since the early 2000s are now in their reproductive age and are already seeking answers with regards to their fertility. OBJECTIVE AND RATIONALE In the light of past decade achievements and observations, this review aims to provide insight into relevant questions for clinicians involved in FP programmes. Have the indications for FP for prepubertal boys changed over time? What is key for patient counselling and ITT sampling based on the latest achievements in animals and research performed with human ITT? How far are we from clinical application of methods to restore reproductive capacity with cryostored ITT? SEARCH METHODS An extensive search for articles published in English or French since January 2010 to June 2020 using keywords relevant to the topic of FP for prepubertal boys was made in the MEDLINE database through PubMed. Original articles on fertility preservation with emphasis on those involving prepubertal testicular tissue, as well as comprehensive and systematic reviews were included. Papers with redundancy of information or with an absence of a relevant link for future clinical application were excluded. Papers on alternative sources of stem cells besides SSCs were excluded. OUTCOMES Preliminary follow-up data indicate that around 27% of boys who have undergone testicular sampling as an FP measure have proved azoospermic and must therefore solely rely on their cryostored ITT to ensure biologic parenthood. Auto-transplantation of ITT appears to be the first technique that could enter pilot clinical trials but should be restricted to tissue free of malignant cells. While in vitro spermatogenesis circumvents the risk linked to cancer cell contamination and has led to offspring in mice, complete spermatogenesis has not been achieved with human ITT. However, generation of haploid germ cells paves the way to further studies aimed at completing the final maturation of germ cells and increasing the efficiency of the processes. WIDER IMPLICATIONS Despite all the research done to date, FP for prepubertal boys remains a relatively young field and is often challenging to healthcare providers, patients and parents. As cryopreservation of ITT is now likely to expand further, it is important not only to acknowledge some of the research questions raised on the topic, e.g. the epigenetic and genetic integrity of gametes derived from strategies to restore fertility with banked ITT but also to provide healthcare professionals worldwide with updated knowledge to launch proper multicollaborative care pathways in the field and address clinical issues that will come-up when aiming for the child's best interest.
Collapse
Affiliation(s)
- Christine Wyns
- Andrology lab, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium.,Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Marc Kanbar
- Andrology lab, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium.,Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Maria Grazia Giudice
- Andrology lab, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium.,Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Jonathan Poels
- Andrology lab, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, Brussels, Belgium.,Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
27
|
Tharmalingam MD, Matilionyte G, Wallace WHB, Stukenborg JB, Jahnukainen K, Oliver E, Goriely A, Lane S, Guo J, Cairns B, Jorgensen A, Allen CM, Lopes F, Anderson RA, Spears N, Mitchell RT. Cisplatin and carboplatin result in similar gonadotoxicity in immature human testis with implications for fertility preservation in childhood cancer. BMC Med 2020; 18:374. [PMID: 33272271 PMCID: PMC7716476 DOI: 10.1186/s12916-020-01844-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 11/06/2020] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Clinical studies indicate chemotherapy agents used in childhood cancer treatment regimens may impact future fertility. However, effects of individual agents on prepubertal human testis, necessary to identify later risk, have not been determined. The study aimed to investigate the impact of cisplatin, commonly used in childhood cancer, on immature (foetal and prepubertal) human testicular tissues. Comparison was made with carboplatin, which is used as an alternative to cisplatin in order to reduce toxicity in healthy tissues. METHODS We developed an organotypic culture system combined with xenografting to determine the effect of clinically-relevant exposure to platinum-based chemotherapeutics on human testis. Human foetal and prepubertal testicular tissues were cultured and exposed to cisplatin, carboplatin or vehicle for 24 h, followed by 24-240 h in culture or long-term xenografting. Survival, proliferation and apoptosis of prepubertal germ stem cell populations (gonocytes and spermatogonia), critical for sperm production in adulthood, were quantified. RESULTS Cisplatin exposure resulted in a significant reduction in the total number of germ cells (- 44%, p < 0.0001) in human foetal testis, which involved an initial loss of gonocytes followed by a significant reduction in spermatogonia. This coincided with a reduction (- 70%, p < 0.05) in germ cell proliferation. Cisplatin exposure resulted in similar effects on total germ cell number (including spermatogonial stem cells) in prepubertal human testicular tissues, demonstrating direct relevance to childhood cancer patients. Xenografting of cisplatin-exposed human foetal testicular tissue demonstrated that germ cell loss (- 42%, p < 0.01) persisted at 12 weeks. Comparison between exposures to human-relevant concentrations of cisplatin and carboplatin revealed a very similar degree of germ cell loss at 240 h post-exposure. CONCLUSIONS This is the first demonstration of direct effects of chemotherapy exposure on germ cell populations in human foetal and prepubertal testis, demonstrating platinum-induced loss of all germ cell populations, and similar effects of cisplatin or carboplatin. Furthermore, these experimental approaches can be used to determine the effects of established and novel cancer therapies on the developing testis that will inform fertility counselling and development of strategies to preserve fertility in children with cancer.
Collapse
Affiliation(s)
- Melissa D Tharmalingam
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK
- KK Women's and Children's Hospital, Bukit Timah Rd, 100, Singapore, 229899, Singapore
| | - Gabriele Matilionyte
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK
| | - William H B Wallace
- Edinburgh Royal Hospital for Sick Children, 9 Sciennes Road, Edinburgh, EH9 1LF, Scotland, UK
| | - Jan-Bernd Stukenborg
- NORDFERTIL Research Lab Stockholm, Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Kirsi Jahnukainen
- NORDFERTIL Research Lab Stockholm, Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Division of Haematology-Oncology and Stem Cell Transplantation, Children's Hospital, University of Helsinki, Helsinki University Central Hospital, Helsinki, Finland
| | - Elizabeth Oliver
- NORDFERTIL Research Lab Stockholm, Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Anne Goriely
- Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX39DS, UK
| | - Sheila Lane
- Department of Paediatrics and Child Health, Oxford University Hospitals NHS Foundation Trust, and Nuffield Department of Womens and Reproductive Health, University of Oxford, Oxford, UK
| | - Jingtao Guo
- Section of Andrology, Division of Urology, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT, USA
- Howard Hughes Medical Institute, Department of Oncological Sciences and Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Bradley Cairns
- Howard Hughes Medical Institute, Department of Oncological Sciences and Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Anne Jorgensen
- Department of Growth and Reproduction, Copenhagen University Hospital (Rigshospitalet), Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Caroline M Allen
- Biomedical Sciences, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Federica Lopes
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK
| | - Richard A Anderson
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK
| | - Norah Spears
- Biomedical Sciences, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Rod T Mitchell
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, Scotland, UK.
- Edinburgh Royal Hospital for Sick Children, 9 Sciennes Road, Edinburgh, EH9 1LF, Scotland, UK.
| |
Collapse
|
28
|
Somigliana E, Mangili G, Martinelli F, Noli S, Filippi F, Bergamini A, Bocciolone L, Buonomo B, Peccatori F. Fertility preservation in women with cervical cancer. Crit Rev Oncol Hematol 2020; 154:103092. [PMID: 32896752 DOI: 10.1016/j.critrevonc.2020.103092] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 07/15/2020] [Accepted: 08/17/2020] [Indexed: 12/16/2022] Open
Abstract
Fertility preservation in women with cervical cancer is a demanding but evolving issue. Some remarkable achievements have been reached, in particular the improvement of primary and secondary prevention and the broadening of the indications for conservative surgery up to FIGO 2018 stage IB2. Natural pregnancy rate and the rate of obstetrics complications following conservative approach is satisfactory even if not optimal. On the other hand, the use of classic strategies for fertility preservation such as oocytes or ovarian cortex freezing is extremely limited, being the uterus compromised by treatment in a high proportion of cases. In fact, the availability of uterine surrogacy can play a role in the counseling and the decision-making process. The recent advent of uterus transplantation is fascinating but, at present, cannot be viewed as a realistic solution.
Collapse
Affiliation(s)
- Edgardo Somigliana
- Dept of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy; Obstet-Gynecol Dept, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| | - Giorgia Mangili
- Obstet-Gynecol Dept, San Raffaele Scientific Institute, IRCCS Milan, Italy
| | - Fabio Martinelli
- Gynecologic Oncology Unit, Fondazione IRCCS National Cancer Institute, Milan, Italy
| | - Stefania Noli
- Dept of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy; Fertility and Procreation Unit, Division of Gynaecologic Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Francesca Filippi
- Obstet-Gynecol Dept, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Alice Bergamini
- Obstet-Gynecol Dept, San Raffaele Scientific Institute, IRCCS Milan, Italy
| | - Luca Bocciolone
- Obstet-Gynecol Dept, San Raffaele Scientific Institute, IRCCS Milan, Italy
| | - Barbara Buonomo
- Fertility and Procreation Unit, Division of Gynaecologic Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Fedro Peccatori
- Fertility and Procreation Unit, Division of Gynaecologic Oncology, European Institute of Oncology IRCCS, Milan, Italy
| |
Collapse
|
29
|
Anwar N, Qureshi IZ, Spears N, Lopes F. In vitro administration of sodium arsenite in mouse prepubertal testis induces germ cell loss and apoptosis. Toxicol In Vitro 2020; 67:104924. [PMID: 32599264 DOI: 10.1016/j.tiv.2020.104924] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 12/28/2022]
Abstract
High levels of arsenic contamination in drinking water pose serious health risks in numerous countries. The documentation reporting arsenic toxicity on reproduction and development is increasing, with evidence of arsenic inducing fertility and developmental issues. Nonetheless, the impact of arsenic exposure on the development of the male reproductive system is not fully elucidated. In the present study, we have investigated the direct effects of arsenic on prepubertal mouse testis using an in vitro testicular organ culture system. Culture medium was supplemented with a range of concentrations of sodium arsenite, examining effects of low (0.5 and 1 μM) and high (10, 50, 100 μM) concentrations, in cultures of post-natal day 5 CD1 mouse testis. In vitro exposure of low arsenic concentrations (0.5 or 1 μM) for 6 days did not cause any change in the testicular morphology, germ cells density, or apoptotic marker cleaved caspase 3 (CC3) expression. In contrast, exposure of prepubertal testis to high arsenic concentrations (10, 50 or 100 μM) induced drastic changes: severe destruction of testicular morphology, with loss of seminiferous tubule integrity; a dose-dependent decrease in germ cell density, and a hundred-fold increase in CC3 expression after 50 μM arsenic exposure. In conclusion, high arsenic treatment induced a dose-dependent induction of apoptosis and germ cell loss in prepubertal mouse testis.
Collapse
Affiliation(s)
- Naureen Anwar
- Quaid-i-Azam University, Department of Animal Sciences, Laboratory of Animal and Human Physiology, PO Box 45320, Islamabad, Pakistan
| | - Irfan Zia Qureshi
- Quaid-i-Azam University, Department of Animal Sciences, Laboratory of Animal and Human Physiology, PO Box 45320, Islamabad, Pakistan
| | - Norah Spears
- University of Edinburgh, School of Biomedical Sciences, Edinburgh, United Kingdom
| | - Federica Lopes
- University of Edinburgh, School of Biomedical Sciences, Edinburgh, United Kingdom..
| |
Collapse
|