1
|
Di Berardino C, Peserico A, Camerano Spelta Rapini C, Liverani L, Capacchietti G, Russo V, Berardinelli P, Unalan I, Damian-Buda AI, Boccaccini AR, Barboni B. Bioengineered 3D ovarian model for long-term multiple development of preantral follicle: bridging the gap for poly(ε-caprolactone) (PCL)-based scaffold reproductive applications. Reprod Biol Endocrinol 2024; 22:95. [PMID: 39095895 PMCID: PMC11295475 DOI: 10.1186/s12958-024-01266-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Assisted Reproductive Technologies (ARTs) have been validated in human and animal to solve reproductive problems such as infertility, aging, genetic selection/amplification and diseases. The persistent gap in ART biomedical applications lies in recapitulating the early stage of ovarian folliculogenesis, thus providing protocols to drive the large reserve of immature follicles towards the gonadotropin-dependent phase. Tissue engineering is becoming a concrete solution to potentially recapitulate ovarian structure, mostly relying on the use of autologous early follicles on natural or synthetic scaffolds. Based on these premises, the present study has been designed to validate the use of the ovarian bioinspired patterned electrospun fibrous scaffolds fabricated with poly(ε-caprolactone) (PCL) for multiple preantral (PA) follicle development. METHODS PA follicles isolated from lamb ovaries were cultured on PCL scaffold adopting a validated single-follicle protocol (Ctrl) or simulating a multiple-follicle condition by reproducing an artificial ovary engrafted with 5 or 10 PA (AO5PA and AO10PA). The incubations were protracted for 14 and 18 days before assessing scaffold-based microenvironment suitability to assist in vitro folliculogenesis (ivF) and oogenesis at morphological and functional level. RESULTS The ivF outcomes demonstrated that PCL-scaffolds generate an appropriate biomimetic ovarian microenvironment supporting the transition of multiple PA follicles towards early antral (EA) stage by supporting follicle growth and steroidogenic activation. PCL-multiple bioengineering ivF (AO10PA) performed in long term generated, in addition, the greatest percentage of highly specialized gametes by enhancing meiotic competence, large chromatin remodeling and parthenogenetic developmental competence. CONCLUSIONS The study showcased the proof of concept for a next-generation ART use of PCL-patterned scaffold aimed to generate transplantable artificial ovary engrafted with autologous early-stage follicles or to advance ivF technologies holding a 3D bioinspired matrix promoting a physiological long-term multiple PA follicle protocol.
Collapse
Affiliation(s)
- Chiara Di Berardino
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100, Teramo, Italy.
| | - Alessia Peserico
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100, Teramo, Italy
| | - Chiara Camerano Spelta Rapini
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100, Teramo, Italy
| | - Liliana Liverani
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Cauerstraße 6, 91058, Erlangen, Germany
- DGS SpA, Via Paolo di Dono 73, 00142, Rome, Italy
| | - Giulia Capacchietti
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100, Teramo, Italy
| | - Valentina Russo
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100, Teramo, Italy
| | - Paolo Berardinelli
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100, Teramo, Italy
| | - Irem Unalan
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Cauerstraße 6, 91058, Erlangen, Germany
| | - Andrada-Ioana Damian-Buda
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Cauerstraße 6, 91058, Erlangen, Germany
| | - Aldo R Boccaccini
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Cauerstraße 6, 91058, Erlangen, Germany
| | - Barbara Barboni
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100, Teramo, Italy
| |
Collapse
|
2
|
Zha Y, Li Y, Lyu W. Research progress on the prevention and treatment of chemotherapy-induced ovarian damage. Zhejiang Da Xue Xue Bao Yi Xue Ban 2024; 53:288-296. [PMID: 38742393 PMCID: PMC11348697 DOI: 10.3724/zdxbyxb-2023-0495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 04/12/2024] [Indexed: 05/16/2024]
Abstract
Chemotherapy is a main treatment option for malignant tumors, but it may cause various adverse effects, including dysfunction of female endocrine system and fertility. Chemotherapy-induced ovarian damage has been concerned with ovarian preservation but also the prevention and treatment of ovarian dysfunction. In this article, the mechanisms of ovarian injury caused by chemotherapy, including apoptosis of the follicle and supporting cells, follicle "burn out", ovarian stromal and microvascular damage; and influencing factors, including age at diagnosis, initial low pre-treatment anti-Müllerian hormone levels, toxicity, dose and regimen of chemotherapy drugs are reviewed based on the latest research results and clinical practice. The article also discusses measures and frontier therapies for the prevention and treatment of ovarian injury, including the application of gonadotropin releasing hormone agonists or antagonists, tyrosine kinase inhibitors, antioxidants, sphingosine-1-phosphate, ceramide-1-phosphate, mammalian target of rapamycin inhibitors, granulocyte-colony stimulating factor, stem cell therapy and artificial ovaries.
Collapse
Affiliation(s)
- Yuxin Zha
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
| | - Yang Li
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou 310006, China
| | - Weiguo Lyu
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China.
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou 310006, China.
- Zhejiang Provincial Key Laboratory of Women's Reproductive Health, Hangzhou 310006, China.
| |
Collapse
|
3
|
Sistani MN, Zavareh S, Valojerdi MR, Salehnia M. Reconstruction of ovarian follicular-like structure by recellularization of a cell-free human ovarian scaffold with mouse fetal ovarian cells. Cytotechnology 2024; 76:27-38. [PMID: 38304626 PMCID: PMC10828258 DOI: 10.1007/s10616-023-00595-x] [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: 05/26/2023] [Accepted: 09/04/2023] [Indexed: 02/03/2024] Open
Abstract
The present study assessed the supportive roles of the decellularized human ovarian tissue in homing of mouse fetal ovarian cells into the scaffold as well as the formation of the follicular-like structure. The human ovarian cortical tissues were decellularized by three freeze-thaw cycles and then, treated with Triton X-100 for 15 h and 0.5% sodium dodecyl sulfate for 72 h. After isolation and preparation of mouse fetal ovarian cells (19 dpc) they were seeded into the decellularized scaffolds and cultured for 7 days, then using a light microscope, laser confocal scanning microscope, and scanning electron microscope these scaffolds were studied. Analysis of gene expression related to oocyte and follicular cells such as Ddx4, Nobox, Gdf9, and Connexin37 was assessed by real-time RT-PCR and the DDX4 and GDF9 proteins were detected by immunohistochemistry. The result showed that the human ovarian tissue was decellularized properly and the tissue elements and integrity were well preserved. After 7 days of in vitro culture, the fetal ovarian cells attached and penetrated into different sites and depths of the scaffold. The formed organoid within the scaffold showed large round, small polyhedral, and elongated spindle cells similar to the follicle structure. The molecular analysis and immunohistochemistry were confirmed an increase in the expression of genes and proteins related to oocyte and follicular cells in these reconstructed structures. In conclusion, the recellularization of human ovarian scaffolds by mouse fetal ovarian cells could support the follicular-like structure formation and it provides an in vitro model for follicle reconstitution and offers an alternative approach for clinical usage.
Collapse
Affiliation(s)
- Maryam Nezhad Sistani
- Anatomy Department, Faculty of Medical Sciences, Tarbiat Modares University, 14115-111, Tehran, Iran
| | - Saeed Zavareh
- School of Biology, Damghan University, Damghan, Iran
| | | | - Mojdeh Salehnia
- Anatomy Department, Faculty of Medical Sciences, Tarbiat Modares University, 14115-111, Tehran, Iran
| |
Collapse
|
4
|
Khaleghi S, Eivazkhani F, Tavana S, Moini A, Novin MG, Stoyan P, Nazarian H, Fathi R. Follicular reconstruction and neo-oogenesis in xenotransplantation of human ovarian isolated cells derived from chemotherapy-induced POF patients. J Biol Eng 2023; 17:70. [PMID: 37986177 PMCID: PMC10662631 DOI: 10.1186/s13036-023-00384-2] [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: 03/24/2023] [Accepted: 10/17/2023] [Indexed: 11/22/2023] Open
Abstract
BACKGROUND Developing new strategies to restore fertility in patients with chemotherapy-induced Premature Ovarian Failure (Chemo-POF) is important. We aimed to construct an Artificial Ovary (AO) by seeding Human Ovarian Cortical Cells (HOCCs) into Human ovarian Decellularized Cortical Tissue (DCT). We assessed the AO's ability to produce new ovarian follicles following xenotransplantation to NMRI mice. MATERIAL AND METHODS The DCTs were prepared, and cell removal was confirmed through DNA content, MTT assay, DAPI and H&E staining. Next, HOCCs were isolated from both Chemo-POF and Trans (as a control group) ovarian patients. The HOCCs were characterized using immunostaining (FRAGILIS, Vimentin, and Inhibin α) and real time PCR (DDX4, STELLA, FRAGILIS, Vimentin, FSH-R, KI67) assays. The HOCCs were then seeded into the DCTs and cultured for one week to construct an AO, which was subsequently xenotransplanted into the mice. The existence of active follicles within the AO was studied with H&E and immunofluorescence (GDF9) staining, Real-time PCR (GDF9, ZP3) and hormone analysis (Estradiol, FSH and AMH). RESULTS The results of gene expression and immunostaining showed that 85-86% of the isolated cells from both Trans and Chemo-POF groups were positive for vimentin, while 2-5% were granulosa cells and OSCs were less than 3%. After xenotransplantation, histological study confirmed the presence of morphologically healthy reconstructed human ovarian follicles. Additionally, immunofluorescence staining of GDF9 and hormonal assay confirmed the presence of secretory-active follicles on the AO. CONCLUSION Our findings demonstrate that an artificial ovary produced by seeding HOCCs on DCT can support HOCCs proliferation as well as neo-oogenesis, and enable sex hormone secretion following xenotransplantation.
Collapse
Affiliation(s)
- Sara Khaleghi
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farideh Eivazkhani
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Somayeh Tavana
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Ashraf Moini
- Department of Endocrinology and Female Infertility, Royan Institute of Reproductive Biomedicine, ACECR, Tehran, Iran
- Breast Disease Research Center (BDRC), Tehran University of Medical Science, Tehran, Iran
| | - Marefat Ghaffari Novin
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Petkov Stoyan
- Platform Degenerative Diseases, German Primate Center, GmbH, Leibniz Institute for Primate Research, Göttingen, 37077, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, 37077, Germany
| | - Hamid Nazarian
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Rouhollah Fathi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.
| |
Collapse
|
5
|
Canosa S, Revelli A, Gennarelli G, Cormio G, Loizzi V, Arezzo F, Petracca EA, Carosso AR, Cimadomo D, Rienzi L, Vaiarelli A, Ubaldi FM, Silvestris E. Innovative Strategies for Fertility Preservation in Female Cancer Survivors: New Hope from Artificial Ovary Construction and Stem Cell-Derived Neo-Folliculogenesis. Healthcare (Basel) 2023; 11:2748. [PMID: 37893822 PMCID: PMC10606281 DOI: 10.3390/healthcare11202748] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/05/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Recent advances in anticancer treatment have significantly improved the survival rate of young females; unfortunately, in about one third of cancer survivors the risk of ovarian insufficiency and infertility is still quite relevant. As the possibility of becoming a mother after recovery from a juvenile cancer is an important part of the quality of life, several procedures to preserve fertility have been developed: ovarian surgical transposition, induction of ovarian quiescence by gonadotropin-releasing hormone agonists (GnRH-a) treatment, and oocyte and/or ovarian cortical tissue cryopreservation. Ovarian tissue cryostorage and allografting is a valuable technique that applies even to prepubertal girls; however, some patients cannot benefit from it due to the high risk of reintroducing cancer cells during allograft in cases of ovary-metastasizing neoplasias, such as leukemias or NH lymphomas. Innovative techniques are now under investigation, as in the construction of an artificial ovary made of isolated follicles inserted into an artificial matrix scaffold, and the use of stem cells, including ovarian stem cells (OSCs), to obtain neo-folliculogenesis and the development of fertilizable oocytes from the exhausted ovarian tissue. This review synthesizes and discusses these innovative techniques, which potentially represent interesting strategies in oncofertility programs and a new hope for young female cancer survivors.
Collapse
Affiliation(s)
- Stefano Canosa
- IVIRMA, Global Research Alliance, LIVET, 10126 Turin, Italy; (A.R.); (G.G.)
| | - Alberto Revelli
- IVIRMA, Global Research Alliance, LIVET, 10126 Turin, Italy; (A.R.); (G.G.)
- Gynecology and Obstetrics 2U, Department of Surgical Sciences, S. Anna Hospital, University of Turin, 10126 Turin, Italy
| | - Gianluca Gennarelli
- IVIRMA, Global Research Alliance, LIVET, 10126 Turin, Italy; (A.R.); (G.G.)
- Gynecology and Obstetrics 1U, Physiopathology of Reproduction and IVF Unit, Department of Surgical Sciences, S. Anna Hospital, University of Turin, 10126 Turin, Italy;
| | - Gennaro Cormio
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (G.C.); (V.L.); (E.A.P.); (E.S.)
- Department of Interdisciplinary Medicine (DIM), University of Bari “Aldo Moro”, 70121 Bari, Italy
| | - Vera Loizzi
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (G.C.); (V.L.); (E.A.P.); (E.S.)
- Department of Interdisciplinary Medicine (DIM), University of Bari “Aldo Moro”, 70121 Bari, Italy
| | - Francesca Arezzo
- Obstetrics and Gynecology Unit, Department of Biomedical Sciences and Human Oncology, University of “Aldo Moro”, 70124 Bari, Italy
| | - Easter Anna Petracca
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (G.C.); (V.L.); (E.A.P.); (E.S.)
| | - Andrea Roberto Carosso
- Gynecology and Obstetrics 1U, Physiopathology of Reproduction and IVF Unit, Department of Surgical Sciences, S. Anna Hospital, University of Turin, 10126 Turin, Italy;
| | - Danilo Cimadomo
- IVIRMA, Global Research Alliance, GENERA, Clinica Valle Giulia, 00197 Rome, Italy; (D.C.); (L.R.); (A.V.); (F.M.U.)
| | - Laura Rienzi
- IVIRMA, Global Research Alliance, GENERA, Clinica Valle Giulia, 00197 Rome, Italy; (D.C.); (L.R.); (A.V.); (F.M.U.)
- Department of Biomolecular Sciences, University of Urbino “Carlo Bo”, 61029 Urbino, Italy
| | - Alberto Vaiarelli
- IVIRMA, Global Research Alliance, GENERA, Clinica Valle Giulia, 00197 Rome, Italy; (D.C.); (L.R.); (A.V.); (F.M.U.)
| | - Filippo Maria Ubaldi
- IVIRMA, Global Research Alliance, GENERA, Clinica Valle Giulia, 00197 Rome, Italy; (D.C.); (L.R.); (A.V.); (F.M.U.)
| | - Erica Silvestris
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (G.C.); (V.L.); (E.A.P.); (E.S.)
| |
Collapse
|
6
|
Varlas VN, Bors RG, Cretoiu R, Carp-Veliscu A, Mehedintu C, Cirstoiu M. The Artificial Ovary: the Next Step in Fertility Preservation in Cancer Patients. MAEDICA 2023; 18:477-482. [PMID: 38023739 PMCID: PMC10674111 DOI: 10.26574/maedica.2023.18.3.477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2023]
Abstract
The cryopreservation procedure of ovarian tissue is used for subsequent transplantation to preserve fertility in cancer patients. In the case of cancers with possible ovarian damage, due to the increased risk of transmission of malignant cells in the cryopreserved ovarian tissue, after remission of the disease, the transplant cannot be performed due to the high rate of recurrence. Thus, to resolve fertility preservation in these cancer patients, making an artificial ovary that could be transplanted under maximum safety conditions was necessary. This was not easy to achieve because it was essential to develop a porous and rigid matrix that could encapsulate and protect the ovarian follicles and, at the same time, create an optimal neuroendocrine environment. The present article analyzes the technological progress in creating an artificial ovary, the opportunity for transplantation, the proper counseling of these patients, and the prognosis regarding using this modern technique to preserve fertility.
Collapse
Affiliation(s)
- Valentin Nicolae Varlas
- Department of Obstetrics and Gynecology, Filantropia Clinical Hospital, Bucharest, Romania
- "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| | - Roxana Georgiana Bors
- Department of Obstetrics and Gynecology, Filantropia Clinical Hospital, Bucharest, Romania
- Victoria Medical Center, Bucharest, Romania
| | - Rebeca Cretoiu
- Department of Pituitary and Neuroendocrine Disorders, C.I. Parhon National Institute of Endocrinology, Bucharest, Romania
| | - Andreea Carp-Veliscu
- "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
- Department of Obstetrics and Gynecology, "Prof. Dr. Panait Sârbu" Hospital, Bucharest, Romania
- Embryos Fertility Clinic, Bucharest, Romania
| | - Claudia Mehedintu
- Department of Obstetrics and Gynecology, Filantropia Clinical Hospital, Bucharest, Romania
- "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| | - Monica Cirstoiu
- "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
- Department of Obstetrics and Gynecology, University Emergency Hospital Bucharest, Romania
| |
Collapse
|
7
|
Li N, Fan X, Liu L, Liu Y. Therapeutic effects of human umbilical cord mesenchymal stem cell-derived extracellular vesicles on ovarian functions through the PI3K/Akt cascade in mice with premature ovarian failure. Eur J Histochem 2023; 67:3506. [PMID: 37503653 PMCID: PMC10476539 DOI: 10.4081/ejh.2023.3506] [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: 07/28/2022] [Accepted: 12/28/2022] [Indexed: 07/29/2023] Open
Abstract
Premature ovarian failure (POF) mainly refers to ovarian dysfunction in females younger than forty. Mesenchymal stem cells (MSCs) are considered an increasingly promising therapy for POF. This study intended to uncover the therapeutic effects of human umbilical cord MSC-derived extracellular vesicles (hucMSCEVs) on POF. hucMSCs were identified by observing morphology and examining differentiation capabilities. EVs were extracted from hucMSCs and later identified utilizing nanoparticle tracking analysis, transmission electron microscopy, and Western blotting. POF mouse models were established by injecting D-galactose (Dgal). The estrous cycles were assessed through vaginal cytology, and serum levels of follicle-stimulating hormone (FSH), luteinizing hormone (LH), anti-mullerian hormone (AMH), estradiol (E2), and progesterone (P) were measured by ELISA. The human ovarian granulosa cell line KGN was used for in vitro experiments. The uptake of hucMSC-EVs by KGN cells was detected. After D-gal treatment, cell proliferation and apoptosis were assessed via CCK-8 assay and flow cytometry. The PI3K/Akt pathway-related proteins were determined by Western blotting. Our results revealed that POF mice had prolonged estrous cycles, increased FSH and LH levels, and decreased AMH, E2, and P levels. Treatment with hucMSC-EVs partially counteracted the above changes. D-gal treatment reduced proliferation and raised apoptosis in KGN cells, while hucMSC-EV treatment annulled the changes. D-gal-treated cells exhibited downregulated p-PI3K/PI3K and p-Akt/Akt levels, while hucMSC-EVs activated the PI3K/Akt pathway. LY294002 suppressed the roles of hucMSC-EVs in promoting KGN cell proliferation and lowering apoptosis. Collectively, hucMSC-EVs facilitate proliferation and suppress apoptosis of ovarian granulosa cells by activating the PI3K/Akt pathway, thereby alleviating POF.
Collapse
Affiliation(s)
- Nan Li
- Department of Gynecological Ward, The Third Affiliated Hospital, Jinzhou Medical University, Jinzhou.
| | - Xue Fan
- Department of Gynecological Ward, The Third Affiliated Hospital, Jinzhou Medical University, Jinzhou.
| | - Lihong Liu
- Department of Gynecological Ward, The Third Affiliated Hospital, Jinzhou Medical University, Jinzhou.
| | - Yanbing Liu
- Department of Gynecological Ward, The Third Affiliated Hospital, Jinzhou Medical University, Jinzhou.
| |
Collapse
|
8
|
Antonouli S, Di Nisio V, Messini C, Daponte A, Rajender S, Anifandis G. A comprehensive review and update on human fertility cryopreservation methods and tools. Front Vet Sci 2023; 10:1151254. [PMID: 37143497 PMCID: PMC10151698 DOI: 10.3389/fvets.2023.1151254] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 03/29/2023] [Indexed: 05/06/2023] Open
Abstract
The broad conceptualization of fertility preservation and restoration has become already a major concern in the modern western world since a large number of individuals often face it in the everyday life. Driven by different health conditions and/or social reasons, a variety of patients currently rely on routinely and non-routinely applied assisted reproductive technologies, and mostly on the possibility to cryopreserve gametes and/or gonadal tissues for expanding their reproductive lifespan. This review embraces the data present in human-focused literature regarding the up-to-date methodologies and tools contemporarily applied in IVF laboratories' clinical setting of the oocyte, sperm, and embryo cryopreservation and explores the latest news and issues related to the optimization of methods used in ovarian and testicular tissue cryopreservation.
Collapse
Affiliation(s)
- Sevastiani Antonouli
- Department of Clinical Chemistry, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| | - Valentina Di Nisio
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Christina Messini
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larisa, Greece
| | - Alexandros Daponte
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larisa, Greece
| | - Singh Rajender
- Division of Endocrinology, Central Drug Research Institute, Lucknow, India
| | - George Anifandis
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larisa, Greece
| |
Collapse
|
9
|
Morphological and Chemical Investigation of Ovarian Structures in a Bovine Model by Contrast-Enhanced X-ray Imaging and Microscopy. Int J Mol Sci 2023; 24:ijms24043545. [PMID: 36834956 PMCID: PMC9963314 DOI: 10.3390/ijms24043545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
An improved understanding of an ovary's structures is highly desirable to support advances in folliculogenesis knowledge and reproductive medicine, with particular attention to fertility preservation options for prepubertal girls with malignant tumors. Although currently the golden standard for structural analysis is provided by combining histological sections, staining, and visible 2D microscopic inspection, synchrotron radiation phase-contrast microtomography is becoming a new challenge for three-dimensional studies at micrometric resolution. To this aim, the proper use of contrast agents can improve the visualization of internal structures in ovary tissues, which normally present a low radiopacity. In this study, we report a comparison of four staining protocols, based on iodine or tungsten containing agents, applied to bovine ovarian tissues fixed in Bouin's solution. The microtomography (microCT) analyses at two synchrotron facilities under different set-ups were performed at different energies in order to maximize the image contrast. While tungsten-based agents allow large structures to be well identified, Iodine ones better highlight smaller features, especially when acquired above the K-edge energy of the specific metal. Further scans performed at lower energy where the setup was optimized for overall quality and sensitivity from phase-contrast still provided highly resolved visualization of follicular and intrafollicular structures at different maturation stages, independent of the staining protocol. The analyses were complemented by X-ray Fluorescence mapping on 2D sections, showing that the tungsten-based agent has a higher penetration in this type of tissues.
Collapse
|
10
|
del Valle L, Corchón S, Palop J, Rubio JM, Celda L. The experience of female oncological patients and fertility preservation: A phenomenology study. Eur J Cancer Care (Engl) 2022; 31:e13757. [PMID: 36354130 PMCID: PMC9787558 DOI: 10.1111/ecc.13757] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 09/07/2022] [Accepted: 10/09/2022] [Indexed: 11/11/2022]
Abstract
OBJECTIVE The aim of this study is to analyse the experience of fertility preservation among female oncological patients. METHODS A phenomenological study was conducted in an Assisted Human Reproduction Unit. The sample was composed of 14 females of reproductive age diagnosed with a cancer that may affect reproductive function, who had undergone a fertility preservation procedure. In-depth interviews were carried out, and the transcripts were entred into the Atlas.ti.v.8.2.3 qualitative analysis software package. The steps proposed by Taylor-Bogdan were followed in the analysis of the data. RESULTS Four thematic categories emerged in relation to the experience with the fertility preservation process: interruption of life plans and changes in the person; knowledge about fertility preservation options, reproductive capacity and impact because of the possible loss; dyadic relationshipsand social representations of fertility. CONCLUSIONS Patients suffer from difficulties when making decisions about fertility whilst dealing with a cancer diagnosis. They need adequate information and support from health professionals. Despite increasing awareness of fertility preservation, there is a lack of knowledge regarding patients' experiences and needs related to this process.
Collapse
Affiliation(s)
- Lara del Valle
- Nursing DepartmentFaculty of Nursing and Podiatry, University of ValenciaValenciaSpain
| | | | - Josefa Palop
- Nursing DepartmentFaculty of Nursing and Podiatry, University of ValenciaValenciaSpain
| | - Jose María Rubio
- Department of Assisted Human ReproductionAssisted Human Reproduction Unit of La Fe (AHRU)ValenciaSpain
| | - Luis Celda
- Nursing DepartmentFaculty of Nursing and Podiatry, University of ValenciaValenciaSpain
| |
Collapse
|
11
|
Automatic Evaluation for Bioengineering of Human Artificial Ovary: A Model for Fertility Preservation for Prepubertal Female Patients with a Malignant Tumor. Int J Mol Sci 2022; 23:ijms232012419. [PMID: 36293273 PMCID: PMC9604043 DOI: 10.3390/ijms232012419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/30/2022] [Accepted: 10/12/2022] [Indexed: 11/17/2022] Open
Abstract
Introduction: The in vitro culture of primordial follicles is the only available option for preserving fertility in prepubertal girls with malignant tumors. The cultivation of primordial follicles in scaffolds as artificial ovaries is a promising approach for this. Methods: Dissociated follicles were placed into an artificial ovarian scaffold composed of fibrinogen and thrombin. The follicles were cultured in a dish dedicated to live cell imaging and observed for growth using immunofluorescence and development via optical microscopy. The morphology of the follicles in the scaffold was three-dimensionally reconstructed using the Imaris software. Growth and development were also quantified. Results: The morphology of artificial ovaries began to degrade over time. Within approximately 7 days, primordial follicles were activated and grew into secondary follicles. A comparison of optical and confocal microscopy results revealed the superior detection of live cells using confocal microscopy. The three-dimensional reconstruction of the confocal microscopy data enabled the automatic enumeration and evaluation of the overall morphology of many follicles. Conclusions: The novel artificial ovary-enabled primordial follicles to enter the growth cycle after activation and grow into secondary follicles. The use of a fibrin scaffold as a carrier preserves the developmental potential of primordial germ cells and is a potentially effective method for preserving fertility in prepubertal girls.
Collapse
|