1
|
Daddangadi A, Saruparia S, Predheepan D, Uppangala S, Kabekkodu SP, Nadeem Khan G, Kalthur G, Talevi R, Adiga SK. Impact of vitrification conditions on genetic and functional competence of prepubertal mouse oocytes. Reprod Biol 2025; 25:101022. [PMID: 40279869 DOI: 10.1016/j.repbio.2025.101022] [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: 09/29/2024] [Revised: 04/12/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
Oocyte in vitro maturation (IVM) and vitrification are being considered as fertility preservation strategies for prepubertal cancer patients. Since prepubertal oocytes have differential sensitivity and response to vitrification compared to adult oocytes, there is a need to optimize the technique to improve the outcome. This study specifically looked into the effect of varying equilibration time and temperatures on the survival and functional competence of prepubertal mouse oocytes. Germinal vesicle (GV) stage and in vitro matured, metaphase II stage sibling oocytes retrieved from 2-week-old Swiss albino mice were equilibrated at 24 °C and 37 °C for 10 and 15 min during vitrification. GV vitrified-IVM (GVV) and GV IVM-vitrified (MIIV) oocytes that survived post-warming were assessed for mitochondrial potential, spindle integrity, spindle checkpoint transcripts, and DNA integrity. The GVV oocytes equilibrated at 37 °C for 15 min had a significantly lower maturation rate (P < 0.01). Survival was reduced when MIIV oocytes were equilibrated at 37 °C, regardless of equilibration duration (P < 0.05). The meiotic spindle and DNA integrity were affected at 37 °C/15 min equilibration (P < 0.01). IVM prepubertal mouse oocytes are at higher risk of experiencing cryo-damage with 37 °C equilibration. Hence, fertility preservation protocols must be refined and individualized for prepubertal age to safeguard the genetic and functional integrity of such oocytes.
Collapse
Affiliation(s)
- Akshatha Daddangadi
- Centre of Excellence in Clinical Embryology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576 104, India
| | - Siyona Saruparia
- Centre of Excellence in Clinical Embryology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576 104, India
| | - Dhakshanya Predheepan
- Centre of Excellence in Clinical Embryology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576 104, India
| | - Shubhashree Uppangala
- Division of Reproductive Genetics, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576 104, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal, Manipal Academy of Higher Education, Manipal 576 104, India
| | - G Nadeem Khan
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal, Manipal Academy of Higher Education, Manipal 576 104, India
| | - Guruprasad Kalthur
- Division of Reproductive Biology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576 104, India
| | - Riccardo Talevi
- Dipartimento di Biologia, Università di Napoli "Federico II", Complesso Universitario di Monte S Angelo, Napoli, Italy
| | - Satish Kumar Adiga
- Centre of Excellence in Clinical Embryology, Department of Reproductive Science, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576 104, India.
| |
Collapse
|
2
|
Mihajlovic M, Pásztor-Jánoska DK, Cadenas J, Adrados CS, Andersen CY, Kristensen SG, Lind JU. 3D culture of ovarian follicles in granular and nanofibrillar hydrogels. BIOMATERIALS ADVANCES 2024; 164:213987. [PMID: 39128246 DOI: 10.1016/j.bioadv.2024.213987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 06/21/2024] [Accepted: 08/02/2024] [Indexed: 08/13/2024]
Abstract
3D culture of ovarian follicles in hydrogel matrices is an important emerging tool for basic scientific studies as well as clinical applications such as fertility preservation. For optimizing and scaling 3D culture of preantral follicles, there is a need for identifying biomaterial matrices that simplifies and improves the current culture procedures. At present, microencapsulation of follicles in alginate beads is the most commonly used approach. However, this technique involves notable manual handling and is best suited for encapsulation of single or several follicles. As a potential alternative, we here explore the suitability of different particle-based hydrogel matrices, where follicles can easily be introduced in tunable 3D environments, in large numbers. Specifically, we study the growth of secondary murine follicles in microgranular alginate and nanofibrillar cellulose matrices, with and without cell-binding cues, and map follicle growth against the viscoelastic properties of the matrices. We cultured follicles within the particle-based hydrogels for 10 days and continuously monitored their size, survival, and tendency to extrude oocytes. Interestingly, we observed that the diameter of the growing follicles increased significantly in the particle-based matrices, as compared to state-of-the-art alginate micro-encapsulation. On the other hand, the follicles displayed an increased tendency for early oocyte extrusion in the granular matrices, leading to a notable reduction in the number of intact follicles. We propose that this may be caused by impaired diffusion of nutrients and oxygen through thicker matrices, attributable to our experimental setup. Still, our findings suggest that viscoelastic, granular hydrogels represent promising matrices for 3D culture of early-stage ovarian follicles. In particular, these materials may easily be implemented in advanced culturing devices such as micro-perfusion systems.
Collapse
Affiliation(s)
- Marko Mihajlovic
- Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | | | - Jesús Cadenas
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Cristina Subiran Adrados
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, 2100 Copenhagen, Denmark; The Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Claus Yding Andersen
- The Fertility Clinic, Copenhagen University Hospital Herlev, 2730 Herlev, Denmark
| | - Stine Gry Kristensen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Johan Ulrik Lind
- Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.
| |
Collapse
|
3
|
Aminian S, Mazoochi T, Hosseini ES, Jamalzaei P, Taheri MA. Protective Effect of Bio-Scaffold Against Vitrification Damage in Mouse Ovarian Tissue. Reprod Sci 2024; 31:3512-3520. [PMID: 39210236 DOI: 10.1007/s43032-024-01680-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: 04/28/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024]
Abstract
Ovarian tissue cryopreservation is regarded as useful method for fertility preservation. This study aimed to preserve most of the follicular reserve from the destructive effects of cryoprotectant solutions and liquid nitrogen. For this purpose, 48 female NMRI mice (8 weeks old) were randomly divided into six groups: Fresh (not vitrified), Vitrification (not encapsulated), Alginate 1 (encapsulated in 1% alginate hydrogel before placing in vitrification solutions), Alginate 2 (encapsulated in 1% alginate hydrogel before placing in liquid nitrogen), Aloe vera 1 (encapsulated in Aloe vera pieces before placing in vitrification solutions), Aloe vera 2 (encapsulated in Aloe vera pieces before placing in liquid nitrogen). After vitrification and warming, the histological evaluation showed that the average number of intact primordial follicles decreased significantly in all groups compared to the Fresh group. (P < 0.05). Results of evaluating the expression of apoptosis-related genes showed that the ratio of Bax/Bcl2 and P53 significantly decreased in the Alginate 2 group compared with the vitrification group. The level of Kit gene (KIT proto-oncogeni receptor tyrosine kinase gene) expression was either the same or lower in the experimental groups than in the vitrification group, but there was no statistically significant difference. Levels of tissue nitric oxide (NO) and malondialdehyde (MDA) in Alginate groups 1 and 2 showed a significant decrease compared with the vitrification group (P < 0.05). To conclude, Encapsulation of ovaries in 1% alginate hydrogel before immersion in liquid nitrogen may reduce the damage caused by cryopreservation.
Collapse
Affiliation(s)
- Saba Aminian
- Gametogenesis Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Tahereh Mazoochi
- Gametogenesis Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Elahe Seyed Hosseini
- Gametogenesis Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Parisa Jamalzaei
- Department of Anatomical Sciences & Cognitive Neuroscience, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Maryam Akhavan Taheri
- Gametogenesis Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
4
|
Zaniker EJ, Zhang J, Russo D, Huang R, Suritis K, Drake RS, Barlow-Smith E, Shalek AK, Woodruff TK, Xiao S, Goods BA, Duncan FE. Follicle-intrinsic and spatially distinct molecular programs drive follicle rupture and luteinization during ex vivo mammalian ovulation. Commun Biol 2024; 7:1374. [PMID: 39443665 PMCID: PMC11500180 DOI: 10.1038/s42003-024-07074-9] [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: 05/09/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024] Open
Abstract
During ovulation, the apical wall of the preovulatory follicle breaks down to facilitate gamete release. In parallel, the residual follicle wall differentiates into a progesterone-producing corpus luteum. Disruption of ovulation, whether through contraceptive intervention or infertility, has implications for women's health. In this study, we harness the power of an ex vivo ovulation model and machine-learning guided microdissection to identify differences between the ruptured and unruptured sides of the follicle wall. We demonstrate that the unruptured side exhibits clear markers of luteinization after ovulation while the ruptured side exhibits cell death signals. RNA-sequencing of individual follicle sides reveals 2099 differentially expressed genes (DEGs) between follicle sides without ovulation induction, and 1673 DEGs 12 h after induction of ovulation. Our model validates molecular patterns consistent with known ovulation biology even though this process occurs in the absence of the ovarian stroma, vasculature, and immune cells. We further identify previously unappreciated pathways including amino acid transport and Jag-Notch signaling on the ruptured side and glycolysis, metal ion processing, and IL-11 signaling on the unruptured side of the follicle. This study yields key insights into follicle-inherent, spatially-defined pathways that underlie follicle rupture, which may further understanding of ovulation physiology and advance women's health.
Collapse
Affiliation(s)
- Emily J Zaniker
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Jiyang Zhang
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA
| | - Daniela Russo
- Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Broad Institute, Harvard University & Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA
| | - Ruixu Huang
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Kristine Suritis
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Riley S Drake
- Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Broad Institute, Harvard University & Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA
| | | | - Alex K Shalek
- Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Broad Institute, Harvard University & Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA
| | - Teresa K Woodruff
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Department of Obstetrics and Gynecology, Michigan State University, East Lansing, MI, USA
| | - Shuo Xiao
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA
| | - Brittany A Goods
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA.
| | - Francesca E Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
5
|
Ebrahimi M, Dattena M, Mara L, Pasciu V, Sotgiu FD, Chessa F, Luciano AM, Berlinguer F. In vitro production of meiotically competent oocytes from early antral follicles in sheep. Theriogenology 2024; 226:253-262. [PMID: 38950486 DOI: 10.1016/j.theriogenology.2024.06.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/03/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024]
Abstract
The potential of using long in vitro culture (LIVC) of cumulus-oocyte complexes (COCs) from early antral follicles (EAFs) as an assisted reproductive technology in cattle has shown promising results. This study explored the feasibility of applying this technology to sheep as seasonal breeding animals. Ovaries from sheep were collected during both the breeding and non-breeding seasons. COCs were isolated from EAFs (350-450 μm) and cultured in TCM199 medium supplemented with 0.15 μg/mL Zn sulfate, 10-4IU/mL FSH, 10 ng/mL estradiol, 50 ng/mL testosterone, 50 ng/mL progesterone, and 5 μM Cilostamide. After five days of LIVC, the COCs were submitted to an in vitro maturation procedure. The results indicate successful in vitro development of COCs, evidenced by a significant increase in oocyte diameter (p < 0.000) and the preservation of gap junction communication between oocyte and cumulus cells. The gradual uncoupling was accompanied by a progressive chromatin transition from the non-surrounded nucleolus (NSN) to the surrounded nucleolus (SN) (p < 0.000), coupled with a gradual decrease in global transcriptional activity and an increase in oocyte meiotic competence (p < 0.000). Maintenance of oocyte-cumulus investment architecture, viability, and metaphase II capability was significantly higher in COCs collected during the breeding season (p < 0.000), suggesting higher quality than those obtained during the non-breeding season. In conclusion, our study confirms LIVC feasibility in sheep, emphasizing increased effectiveness during the breeding season in isolating higher-quality COCs from EAFs. These findings can influence improving the LIVC system in mammals with seasonal reproduction.
Collapse
Affiliation(s)
- Mohammadreza Ebrahimi
- Department of Veterinary Medicine, University of Sassari, Via Vienna 2, Sassari, Italy; Department of Animal Science, Agricultural Research Agency of Sardinia, 07100, Sassari, Italy.
| | - Maria Dattena
- Department of Animal Science, Agricultural Research Agency of Sardinia, 07100, Sassari, Italy
| | - Laura Mara
- Department of Animal Science, Agricultural Research Agency of Sardinia, 07100, Sassari, Italy.
| | - Valeria Pasciu
- Department of Veterinary Medicine, University of Sassari, Via Vienna 2, Sassari, Italy
| | - Francesca D Sotgiu
- Department of Veterinary Medicine, University of Sassari, Via Vienna 2, Sassari, Italy
| | - Fabrizio Chessa
- Department of Animal Science, Agricultural Research Agency of Sardinia, 07100, Sassari, Italy
| | - Alberto Maria Luciano
- Reproductive and Developmental Biology Laboratory (ReDBioLab), Department of Veterinary Medicine and Animal Sciences, University of Milan, Via Dell'Università, 6 - 26900, Lodi, Italy
| | - Fiammetta Berlinguer
- Department of Veterinary Medicine, University of Sassari, Via Vienna 2, Sassari, Italy
| |
Collapse
|
6
|
Wang H, Huang Z, Shen X, Lee Y, Song X, Shu C, Wu LH, Pakkiri LS, Lim PL, Zhang X, Drum CL, Zhu J, Li R. Rejuvenation of aged oocyte through exposure to young follicular microenvironment. NATURE AGING 2024; 4:1194-1210. [PMID: 39251866 DOI: 10.1038/s43587-024-00697-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 07/30/2024] [Indexed: 09/11/2024]
Abstract
Reproductive aging is a major cause of fertility decline, attributed to decreased oocyte quantity and developmental potential. A possible cause is aging of the surrounding follicular somatic cells that support oocyte growth and development by providing nutrients and regulatory factors. Here, by creating chimeric follicles, whereby an oocyte from one follicle was transplanted into and cultured within another follicle whose native oocyte was removed, we show that young oocytes cultured in aged follicles exhibited impeded meiotic maturation and developmental potential, whereas aged oocytes cultured within young follicles were significantly improved in rates of maturation, blastocyst formation and live birth after in vitro fertilization and embryo implantation. This rejuvenation of aged oocytes was associated with enhanced interaction with somatic cells, transcriptomic and metabolomic remodeling, improved mitochondrial function and higher fidelity of meiotic chromosome segregation. These findings provide the basis for a future follicular somatic cell-based therapy to treat female infertility.
Collapse
Affiliation(s)
- HaiYang Wang
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore.
| | - Zhongwei Huang
- NUS Bia Echo Asia Centre for Reproductive Longevity and Equality, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xingyu Shen
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Yaelim Lee
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - XinJie Song
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Chang Shu
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Lik Hang Wu
- Cardiovascular Research Institute, National University Health System, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Leroy Sivappiragasam Pakkiri
- Cardiovascular Research Institute, National University Health System, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Poh Leong Lim
- Cardiovascular Research Institute, National University Health System, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xi Zhang
- Center for Cell Dynamics and Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chester Lee Drum
- Cardiovascular Research Institute, National University Health System, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jin Zhu
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Rong Li
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore.
- Center for Cell Dynamics and Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
7
|
Zaniker EJ, Hashim PH, Gauthier S, Ankrum JA, Campo H, Duncan FE. Three-Dimensionally Printed Agarose Micromold Supports Scaffold-Free Mouse Ex Vivo Follicle Growth, Ovulation, and Luteinization. Bioengineering (Basel) 2024; 11:719. [PMID: 39061801 PMCID: PMC11274170 DOI: 10.3390/bioengineering11070719] [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: 06/06/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Ex vivo follicle growth is an essential tool, enabling interrogation of folliculogenesis, ovulation, and luteinization. Though significant advancements have been made, existing follicle culture strategies can be technically challenging and laborious. In this study, we advanced the field through development of a custom agarose micromold, which enables scaffold-free follicle culture. We established an accessible and economical manufacturing method using 3D printing and silicone molding that generates biocompatible hydrogel molds without the risk of cytotoxicity from leachates. Each mold supports simultaneous culture of multiple multilayer secondary follicles in a single focal plane, allowing for constant timelapse monitoring and automated analysis. Mouse follicles cultured using this novel system exhibit significantly improved growth and ovulation outcomes with comparable survival, oocyte maturation, and hormone production profiles as established three-dimensional encapsulated in vitro follicle growth (eIVFG) systems. Additionally, follicles recapitulated aspects of in vivo ovulation physiology with respect to their architecture and spatial polarization, which has not been observed in eIVFG systems. This system offers simplicity, scalability, integration with morphokinetic analyses of follicle growth and ovulation, and compatibility with existing microphysiological platforms. This culture strategy has implications for fundamental follicle biology, fertility preservation strategies, reproductive toxicology, and contraceptive drug discovery.
Collapse
Affiliation(s)
- Emily J. Zaniker
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (E.J.Z.); (P.H.H.); (S.G.)
| | - Prianka H. Hashim
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (E.J.Z.); (P.H.H.); (S.G.)
| | - Samuel Gauthier
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (E.J.Z.); (P.H.H.); (S.G.)
| | - James A. Ankrum
- Roy J. Carver Department of Biomedical Engineering, Pappajohn Biomedical Institute, University of Iowa, Iowa City, IA 52245, USA;
| | - Hannes Campo
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (E.J.Z.); (P.H.H.); (S.G.)
| | - Francesca E. Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (E.J.Z.); (P.H.H.); (S.G.)
| |
Collapse
|
8
|
Zaniker EJ, Zhang M, Hughes L, La Follette L, Atazhanova T, Trofimchuk A, Babayev E, Duncan FE. Shear wave elastography to assess stiffness of the human ovary and other reproductive tissues across the reproductive lifespan in health and disease†. Biol Reprod 2024; 110:1100-1114. [PMID: 38609185 PMCID: PMC11180622 DOI: 10.1093/biolre/ioae050] [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: 01/05/2024] [Revised: 03/15/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
The ovary is one of the first organs to show overt signs of aging in the human body, and ovarian aging is associated with a loss of gamete quality and quantity. The age-dependent decline in ovarian function contributes to infertility and an altered endocrine milieu, which has ramifications for overall health. The aging ovarian microenvironment becomes fibro-inflammatory and stiff with age, and this has implications for ovarian physiology and pathology, including follicle growth, gamete quality, ovulation dynamics, and ovarian cancer. Thus, developing a non-invasive tool to measure and monitor the stiffness of the human ovary would represent a major advance for female reproductive health and longevity. Shear wave elastography is a quantitative ultrasound imaging method for evaluation of soft tissue stiffness. Shear wave elastography has been used clinically in assessment of liver fibrosis and characterization of tendinopathies and various neoplasms in thyroid, breast, prostate, and lymph nodes as a non-invasive diagnostic and prognostic tool. In this study, we review the underlying principles of shear wave elastography and its current clinical uses outside the reproductive tract as well as its successful application of shear wave elastography to reproductive tissues, including the uterus and cervix. We also describe an emerging use of this technology in evaluation of human ovarian stiffness via transvaginal ultrasound. Establishing ovarian stiffness as a clinical biomarker of ovarian aging may have implications for predicting the ovarian reserve and outcomes of Assisted Reproductive Technologies as well as for the assessment of the efficacy of emerging therapeutics to extend reproductive longevity. This parameter may also have broad relevance in other conditions where ovarian stiffness and fibrosis may be implicated, such as polycystic ovarian syndrome, late off target effects of chemotherapy and radiation, premature ovarian insufficiency, conditions of differences of sexual development, and ovarian cancer. Summary sentence: Shear Wave Elastography is a non-invasive technique to study human tissue stiffness, and here we review its clinical applications and implications for reproductive health and disease.
Collapse
Affiliation(s)
- Emily J Zaniker
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Man Zhang
- Department of Radiology, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Lydia Hughes
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Tomiris Atazhanova
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Alexis Trofimchuk
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Elnur Babayev
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Francesca E Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Center for Reproductive Longevity and Equality, Buck Institute for Research on Aging, Novato, CA, USA
| |
Collapse
|
9
|
Pietroforte S, Plough M, Amargant F. Age-associated increased stiffness of the ovarian microenvironment impairs follicle development and oocyte quality and rapidly alters follicle gene expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.09.598134. [PMID: 38915651 PMCID: PMC11195110 DOI: 10.1101/2024.06.09.598134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
In humans, aging triggers cellular and tissue deterioration, and the female reproductive system is the first to show signs of decline. Reproductive aging is associated with decreased ovarian reserve, decreased quality of the remaining oocytes, and decreased production of the ovarian hormones estrogen and progesterone. With aging, both mouse and human ovaries become pro-fibrotic and stiff. However, whether stiffness directly impairs ovarian function, folliculogenesis, and oocyte quality is unknown. To answer this question, we cultured mouse follicles in alginate gels that mimicked the stiffness of reproductively young and old ovaries. Follicles cultured in stiff hydrogels exhibited decreased survival and growth, decreased granulosa cell viability and estradiol synthesis, and decreased oocyte quality. We also observed a reduction in the number of granulosa cell-oocyte transzonal projections. RNA sequencing revealed early changes in the follicle transcriptome in response to stiffness. Follicles cultured in a stiff environment had lower expression of genes related to follicle development and greater expression of genes related to inflammation and extracellular matrix remodeling than follicles cultured in a soft environment. Altogether, our findings suggest that ovarian stiffness directly modulates folliculogenesis and contributes to the progressive decline in oocyte quantity and quality observed in women of advanced maternal age.
Collapse
Affiliation(s)
- Sara Pietroforte
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, USA
| | - Makenzie Plough
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, USA
| | - Farners Amargant
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
10
|
Qiu P, Zhang Y, Lv M, Wang L, Shi D, Luo C. Establishing a 3D-cultured system based on alginate-hydrogel embedding benefits the in vitro maturation of porcine Oocytes. Theriogenology 2024; 225:33-42. [PMID: 38788627 DOI: 10.1016/j.theriogenology.2024.05.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 05/19/2024] [Accepted: 05/19/2024] [Indexed: 05/26/2024]
Abstract
The in vitro maturation (IVM) quality of oocytes is directly related to the subsequent developmental potential of embryos and a fundamental of in vitro embryo production. However, conventional IVM methods fail to maintain the gap-junction intercellular communication (GJIC) between cumulus-oocyte complexes (COCs), which leads to insufficient oocyte maturation. Herein, we investigated the effects of three different three-dimensional (3D) culture methods on oocyte development in vitro, optimized of the alginate-hydrogel embedding method, and assessed the effects of the alginate-hydrogel embedding method on subsequent embryonic developmental potential of oocytes after IVM and parthenogenetic activation (PA). The results showed that Matrigel embedding and alginate-hydrogel embedding benefited the embryonic developmental potential of oocytes after IVM and PA. With the further optimization of alginate-hydrogel embedding, including crosslinking and decrosslinking of parameters, we established a 3D culture system that can significantly increase oocyte maturation and the blastocyst rate of embryos after PA (27.2 ± 1.5 vs 36.7 ± 2.8, P < 0.05). This 3D culture system produced oocytes with markedly increased mitochondrial intensity and membrane potential, which reduced the abnormalities of spindle formation and cortical granule distribution. The alginate-hydrogel embedding system can also remarkably enhance the GJIC between COCs. In summary, based on alginate-hydrogel embedding, we established a 3D culture system that can improve the IVM quality of porcine oocytes, possibly by enhancing GJIC.
Collapse
Affiliation(s)
- Peng Qiu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi Key Laboratory of Animal Breeding & Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, 75 Xiuling Road, Nanning, 530005, China
| | - Yunchuan Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi Key Laboratory of Animal Breeding & Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, 75 Xiuling Road, Nanning, 530005, China
| | - Meiyun Lv
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi Key Laboratory of Animal Breeding & Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, 75 Xiuling Road, Nanning, 530005, China
| | - Lei Wang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi Key Laboratory of Animal Breeding & Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, 75 Xiuling Road, Nanning, 530005, China
| | - Deshun Shi
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi Key Laboratory of Animal Breeding & Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, 75 Xiuling Road, Nanning, 530005, China.
| | - Chan Luo
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi Key Laboratory of Animal Breeding & Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, 75 Xiuling Road, Nanning, 530005, China.
| |
Collapse
|
11
|
Deligiannis SP, Kask K, Modhukur V, Boskovic N, Ivask M, Jaakma Ü, Damdimopoulou P, Tuuri T, Velthut-Meikas A, Salumets A. Investigating the impact of vitrification on bovine ovarian tissue morphology, follicle survival, and transcriptomic signature. J Assist Reprod Genet 2024; 41:1035-1055. [PMID: 38358432 PMCID: PMC11052753 DOI: 10.1007/s10815-024-03038-4] [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: 05/31/2023] [Accepted: 01/18/2024] [Indexed: 02/16/2024] Open
Abstract
PURPOSE Ovarian tissue cryopreservation is vital for fertility preservation, yet its effect on ovarian tissue follicle survival and transcriptomic signature requires further investigation. This study delves into the effects of vitrification on tissue morphology, function, and transcriptomic changes, helping to find possibilities for vitrification protocol improvements. METHODS Ovarian cortex from 19 bovine animals were used to conduct pre- and post-vitrification culture followed by histological assessment, immunohistochemistry, and TUNEL assay. Follicles' functionality was assessed for viability and growth within the tissue and in isolated cultures. RNA-sequencing of ovarian tissue was used to explore the transcriptomic alterations caused by vitrification. RESULTS Follicle density, cell proliferation, and DNA damage in ovarian stroma were unaffected by vitrification. However, vitrified cultured tissue exhibited reduced follicle density of primordial/primary and antral follicles, while freshly cultured tissue manifested reduction of antral follicles. Increased stromal cell proliferation and DNA damage occurred in both groups post-culture. Isolated follicles from vitrified tissue exhibited similar viability to fresh follicles until day 4, after which the survival dropped. RNA-sequencing revealed minor effects of vitrification on transcriptomic signatures, while culture induced significant gene expression changes in both groups. The altered expression of WNT and hormonal regulation pathway genes post-vitrification suggests the molecular targets for vitrification protocol refinement. CONCLUSION Vitrification minimally affects tissue morphology, follicle density, and transcriptomic signature post-thawing. However, culture revealed notable changes in vitrified tissue samples, including reduced follicle density, decreased isolated follicle survival, and alteration in WNT signalling and ovarian hormonal regulation pathways, highlighted them as possible limitations of the current vitrification protocol.
Collapse
Affiliation(s)
- Spyridon P Deligiannis
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, 14186, Stockholm, Sweden.
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, 14186, Stockholm, Sweden.
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, 50406, Tartu, Estonia.
- Department of Obstetrics and Gynecology, University of Helsinki, 00290, Helsinki, Finland.
| | - Keiu Kask
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, 50406, Tartu, Estonia
- Competence Centre of Health Technologies, 50411, Tartu, Estonia
| | - Vijayachitra Modhukur
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, 50406, Tartu, Estonia
- Competence Centre of Health Technologies, 50411, Tartu, Estonia
| | - Nina Boskovic
- Department of Obstetrics and Gynecology, University of Helsinki, 00290, Helsinki, Finland
- Department of Biosciences and Nutrition, Karolinska Institutet, 14183, Huddinge, Sweden
| | - Marilin Ivask
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411, Tartu, Estonia
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, 51014, Tartu, Estonia
| | - Ülle Jaakma
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, 51014, Tartu, Estonia
| | - Pauliina Damdimopoulou
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, 14186, Stockholm, Sweden
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, 14186, Stockholm, Sweden
| | - Timo Tuuri
- Department of Obstetrics and Gynecology, University of Helsinki, 00290, Helsinki, Finland
| | - Agne Velthut-Meikas
- Department of Chemistry and Biotechnology, Tallinn University of Technology, 12618, Tallinn, Estonia
| | - Andres Salumets
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, 14186, Stockholm, Sweden.
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, 14186, Stockholm, Sweden.
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, 50406, Tartu, Estonia.
- Competence Centre of Health Technologies, 50411, Tartu, Estonia.
| |
Collapse
|
12
|
Subiran Adrados C, Cadenas J, Polat SL, Tjäder AS, Blanche P, Kristensen SG. Exploring the potential use of platelet rich plasma (PRP) from adult and umbilical cord blood in murine follicle culture. Reprod Biol 2024; 24:100851. [PMID: 38237503 DOI: 10.1016/j.repbio.2023.100851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/01/2023] [Accepted: 12/27/2023] [Indexed: 04/02/2024]
Abstract
Ovarian follicle culture is a powerful tool to study follicular physiology and has potential applications in clinical and commercial settings. Despite remarkable progress, recreating folliculogenesis in vitro remains challenging for many mammalian species. This study investigates the impact of platelet-rich plasma (PRP) derived from adult blood (human platelet lysate, hPL) and umbilical cord blood (Umbilical cord plasma, UCP) on murine pre-antral follicle culture and oocyte maturation. Pre-antral follicles were cultured individually for 10 days with fetal bovine serum (FBS) serving as the control and two PRP sources (hPL and UCP) and their activated forms (Ac-hPL and Ac-UCP). The results suggest that neither hPL nor UCP, regardless of activation status, improved follicle culture outcomes compared to FBS. Interestingly, activation did not significantly impact the main functional outcomes such as maturation rates, survival, and growth. Oestradiol secretion and oocyte diameter, often considered hallmarks of follicle quality, did not show significant differences between matured and non-matured oocytes across the treatment groups. However, gene expression analysis revealed a significant upregulation of Gdf-9 and Bmp-15 mRNA levels in oocytes from the Ac-UCP group, regardless of maturation stage, suggesting that the accumulation of the mRNA could be due to potential challenges in translation in the Ac-UCP group. In conclusion, this study challenges the hypothesis that PRP, as a serum source, could improve follicle culture outcomes compared to FBS, the gold standard in murine follicle culture. Further research is needed to understand the species-specific effects of PRP and explore other potential factors affecting follicle culture and oocyte quality.
Collapse
Affiliation(s)
- Cristina Subiran Adrados
- Laboratory of Reproductive Biology, Department of Fertility, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, Blegdamsvej 9, DK-2100 Copenhagen, Denmark; Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Jesús Cadenas
- Laboratory of Reproductive Biology, Department of Fertility, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Sofie Lund Polat
- Laboratory of Reproductive Biology, Department of Fertility, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Anna Sanderhage Tjäder
- Laboratory of Reproductive Biology, Department of Fertility, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Paul Blanche
- Department of Biostatistics, University of Copenhagen, Øster Farimagsgade 5, Entrance B, 2nd floor, 1014 Copenhagen, Denmark
| | - Stine Gry Kristensen
- Laboratory of Reproductive Biology, Department of Fertility, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| |
Collapse
|
13
|
Malo C, Oliván S, Ochoa I, Shikanov A. In Vitro Growth of Human Follicles: Current and Future Perspectives. Int J Mol Sci 2024; 25:1510. [PMID: 38338788 PMCID: PMC10855051 DOI: 10.3390/ijms25031510] [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: 12/24/2023] [Revised: 01/21/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
Ovarian tissue cryopreservation is gaining importance as a successful method to restore fertility to girls and young women at high risk of sterility. However, there are concerns regarding the safety of transplantation after ovarian tissue cryopreservation due to the high risk of reintroducing cancer cells and causing disease recurrence. In these cases, the development of culture systems that support oocyte development from the primordial follicle stage is required. Notable achievements have been reached in human follicle in vitro growth in the past decade. Currently, systems for the in vitro culture of ovarian tissue are based on two-dimensional substrates that do not support the survival of follicles or recapitulate the mechanical heterogenicity in the mammalian ovary. Recognition of the importance of special arrangements between cells has spurred research in three-dimensional culture systems, and the provision of a precise culture system that maximizes the diffusion of nutrients and gases through the follicles has raised interest in advanced biomimetic models. The current review critically examines various culture systems employed for the in vitro development of follicles, with a particular focus on solutions utilizing Organ-on-a-Chip (OOC) technology. The emphasis on OOC technology underscores its role as a promising avenue in ensuring the successful cultivation and maintenance of follicular structures during the culture period.
Collapse
Affiliation(s)
- Clara Malo
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; (S.O.); (I.O.)
- Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain
| | - Sara Oliván
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; (S.O.); (I.O.)
- Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain
| | - Ignacio Ochoa
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain; (S.O.); (I.O.)
- Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 50018 Zaragoza, Spain
| | - Ariella Shikanov
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA;
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109, USA
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
14
|
Shen L, Liu J, Luo A, Wang S. The stromal microenvironment and ovarian aging: mechanisms and therapeutic opportunities. J Ovarian Res 2023; 16:237. [PMID: 38093329 PMCID: PMC10717903 DOI: 10.1186/s13048-023-01300-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 10/18/2023] [Indexed: 12/17/2023] Open
Abstract
For decades, most studies of ovarian aging have focused on its functional units, known as follicles, which include oocytes and granulosa cells. However, in the ovarian stroma, there are a variety of somatic components that bridge the gap between general aging and ovarian senescence. Physiologically, general cell types, microvascular structures, extracellular matrix, and intercellular molecules affect folliculogenesis and corpus luteum physiology alongside the ovarian cycle. As a result of damage caused by age-related metabolite accumulation and external insults, the microenvironment of stromal cells is progressively remodeled, thus inevitably perturbing ovarian physiology. With the established platforms for follicle cryopreservation and in vitro maturation and the development of organoid research, it is desirable to develop strategies to improve the microenvironment of the follicle by targeting the perifollicular environment. In this review, we summarize the role of stromal components in ovarian aging, describing their age-related alterations and associated effects. Moreover, we list some potential techniques that may mitigate ovarian aging based on their effect on the stromal microenvironment.
Collapse
Affiliation(s)
- Lu Shen
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Junfeng Liu
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Aiyue Luo
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Shixuan Wang
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
15
|
Khunmanee S, Yoo J, Lee JR, Lee J, Park H. Thiol-yne click crosslink hyaluronic acid/chitosan hydrogel for three-dimensional in vitro follicle development. Mater Today Bio 2023; 23:100867. [PMID: 38179228 PMCID: PMC10765241 DOI: 10.1016/j.mtbio.2023.100867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 11/09/2023] [Accepted: 11/14/2023] [Indexed: 01/06/2024] Open
Abstract
There is a great deal of potential for in vitro follicle growth to provide an alternative approach to fertility preservation. This strategy reduces the possibility of cancer cells re-exposure after transplantation, and it does not require hormone stimulation. Adopting a three-dimensional (3D) culture method helps preserve the architecture of the follicle and promotes the maturity of oocytes. In order to maintain follicle morphology, enhance the quality of mature oocytes, and facilitate meiotic spindle assembly, the current work aimed to develop the 3D in vitro preantral mouse follicle culture method. Thiolated chitosan-co-thiolated hyaluronic (CSHS) hydrogel was designed to evaluate the effects of biomaterials on ovarian follicle development. Isolated follicles from mouse ovaries were randomly divided into alginate (Alg) as a 3D control, thiolated hyaluronic acid (HASH), and CSHS groups. Single follicle was encapsulated in each hydrogel, and performed for 10 days and subsequently ovulated to retrieve mature oocytes on day 11. CSHS hydrogel promoted follicle survival and oocyte viability with maintained spherical morphology of follicle. Matured oocytes with normal appearance of meiotic spindle and chromosome alignment were higher in the CSHS group compared with those in the Alg and HASH groups. Furthermore, CSHS increased expression level of folliculogenesis genes (TGFβ-1, GDF-9) and endocrine-related genes (LHCGR, and FSHR). With various experimental setups and clinical applications, this platform could be applied as an alternative method to in vitro follicle culture with different experimental designs and clinical applications in the long-term period.
Collapse
Affiliation(s)
- Sureerat Khunmanee
- Department of Integrative Engineering, Chung-Ang University, 221 Heukseok-Dong, Dongjak-Gu, Seoul, 06974, Republic of Korea
| | - Jungyoung Yoo
- Department of Biomedical Laboratory Science, Eulji University, Gyeonggi-do, 13135, Republic of Korea
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, 13620, Republic of Korea
| | - Jung Ryeol Lee
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do, 13620, Republic of Korea
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Jaewang Lee
- Department of Biomedical Laboratory Science, Eulji University, Gyeonggi-do, 13135, Republic of Korea
| | - Hansoo Park
- Department of Integrative Engineering, Chung-Ang University, 221 Heukseok-Dong, Dongjak-Gu, Seoul, 06974, Republic of Korea
| |
Collapse
|
16
|
Telfer EE, Grosbois J, Odey YL, Rosario R, Anderson RA. Making a good egg: human oocyte health, aging, and in vitro development. Physiol Rev 2023; 103:2623-2677. [PMID: 37171807 PMCID: PMC10625843 DOI: 10.1152/physrev.00032.2022] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 05/03/2023] [Accepted: 05/06/2023] [Indexed: 05/13/2023] Open
Abstract
Mammalian eggs (oocytes) are formed during fetal life and establish associations with somatic cells to form primordial follicles that create a store of germ cells (the primordial pool). The size of this pool is influenced by key events during the formation of germ cells and by factors that influence the subsequent activation of follicle growth. These regulatory pathways must ensure that the reserve of oocytes within primordial follicles in humans lasts for up to 50 years, yet only approximately 0.1% will ever be ovulated with the rest undergoing degeneration. This review outlines the mechanisms and regulatory pathways that govern the processes of oocyte and follicle formation and later growth, within the ovarian stroma, through to ovulation with particular reference to human oocytes/follicles. In addition, the effects of aging on female reproductive capacity through changes in oocyte number and quality are emphasized, with both the cellular mechanisms and clinical implications discussed. Finally, the details of current developments in culture systems that support all stages of follicle growth to generate mature oocytes in vitro and emerging prospects for making new oocytes from stem cells are outlined.
Collapse
Affiliation(s)
- Evelyn E Telfer
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Johanne Grosbois
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Yvonne L Odey
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Roseanne Rosario
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Richard A Anderson
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
17
|
Sheikh S, Lo BKM, Kaune H, Bansal J, Deleva A, Williams SA. Rescue of follicle development after oocyte-induced ovary dysfunction and infertility in a model of POI. Front Cell Dev Biol 2023; 11:1202411. [PMID: 37614224 PMCID: PMC10443433 DOI: 10.3389/fcell.2023.1202411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 07/20/2023] [Indexed: 08/25/2023] Open
Abstract
The mechanisms and aetiology underlying the development of premature ovarian insufficiency (POI) are poorly understood. However, the oocyte clearly has a role as demonstrated by the Double Mutant (DM) mouse model where ovarian dysfunction (6 weeks) is followed by POI (3 months) due to oocyte-specific deletion of complex and hybrid N- and O-glycans. The ovaries of DM mice contain more primary follicles (3a stage) accompanied by fewer developing follicles, indicating a block in follicle development. To investigate this block, we first analysed early follicle development in postnatal (8-day), pre-pubertal (3-week) and post-pubertal (6-week and 3-month) DM (C1galt1 F/F Mgat1 F/F:ZP3Cre) and Control (C1galt1 F/F Mgat1 F/F) mice. Second, we investigated if transplantation of DM ovaries into a "normal" endocrine environment would restore follicle development. Third, we determined if replacing DM ovarian somatic cells would rescue development of DM oocytes. At 3-week, DM primary 3a follicles contain large oocytes accompanied by early development of a second GC layer and increased GC proliferation. At 6-week, DM primary 3a follicles contain abnormally large oocytes, accompanied with decreased GC proliferation. Transplantation of DM ovaries into a 'normal' endocrine environment did not restore normal follicle development. However, replacing somatic cells by generating reaggregated ovaries (ROs) did enable follicle development to progress and thus highlighted intra-ovarian factors were responsible for the onset of POI in DM females. Thus, these studies demonstrate oocyte-initiated altered communication between GCs and oocytes results in abnormal primary follicles which fail to progress and leads to POI.
Collapse
Affiliation(s)
| | | | | | | | | | - Suzannah A. Williams
- Nuffield Department of Women’s and Reproductive Health, Women’s Centre, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
18
|
Zheng M, Cadenas J, Pors SE, Esa T, Kristensen SG, Mamsen LS, Adrados CS, Andersen CY. Reducing 3D Hydrogel Stiffness, Addition of Oestradiol in a Physiological Concentration and Increasing FSH Concentration Improve In Vitro Growth of Murine Preantral Follicles. Int J Mol Sci 2023; 24:12499. [PMID: 37569872 PMCID: PMC10419395 DOI: 10.3390/ijms241512499] [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: 06/30/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023] Open
Abstract
This study aimed to optimise culture conditions for murine preantral follicles to improve their growth and survival. Preantral follicles (diameter 100-130 µm) were isolated from prepubertal NMRI mice and individually cultured within alginate beads for 12 days. Three conditions were evaluated: (1) follicle re-encapsulation on day 6 of culture-reducing alginate concentration (0.5% to 0.25% w/v), (2) the presence of oestradiol (E2), and (3) increased follicle-stimulating hormone (FSH) concentration in the culture medium (from 10 to 100 mIU/mL FSH). Follicle morphology and growth, as well as anti-Müllerian hormone (AMH) production, were evaluated. From day 8, re-embedded follicles had a larger average diameter compared to follicles without alginate re-encapsulation (0.5% and 0.25% groups, p < 0.05). Oestradiol (1 µM) had a significantly positive effect on the mean follicular diameter and antrum formation (p < 0.001). Moreover, follicles cultured with 100 mIU/mL FSH showed faster growth (p < 0.05) and significantly higher antrum formation (p < 0.05) compared to the low FSH group. Nevertheless, AMH production was not affected by any of the culture conditions. In conclusion, the growth and survival of mouse preantral follicles during a 12-day period were improved by altering the alginate concentration midways during culture and adding E2 and FSH to the culture medium.
Collapse
Affiliation(s)
- Mengxue Zheng
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, 2100 Copenhagen, Denmark; (M.Z.); (J.C.); (S.E.P.); (S.G.K.); (L.S.M.); (C.S.A.)
- The Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Jesús Cadenas
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, 2100 Copenhagen, Denmark; (M.Z.); (J.C.); (S.E.P.); (S.G.K.); (L.S.M.); (C.S.A.)
| | - Susanne Elisabeth Pors
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, 2100 Copenhagen, Denmark; (M.Z.); (J.C.); (S.E.P.); (S.G.K.); (L.S.M.); (C.S.A.)
| | - Tasnim Esa
- The Department of Biotechnology and Biomedicine, Technical University of Denmark, 2800 Kongens Lyngby, Denmark;
| | - Stine Gry Kristensen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, 2100 Copenhagen, Denmark; (M.Z.); (J.C.); (S.E.P.); (S.G.K.); (L.S.M.); (C.S.A.)
| | - Linn Salto Mamsen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, 2100 Copenhagen, Denmark; (M.Z.); (J.C.); (S.E.P.); (S.G.K.); (L.S.M.); (C.S.A.)
| | - Cristina Subiran Adrados
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, 2100 Copenhagen, Denmark; (M.Z.); (J.C.); (S.E.P.); (S.G.K.); (L.S.M.); (C.S.A.)
- The Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Claus Yding Andersen
- The Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
19
|
Ye M, Shan Y, Lu B, Luo H, Li B, Zhang Y, Wang Z, Guo Y, Ouyang L, Gu J, Xiong Z, Zhang T. Creating a semi-opened micro-cavity ovary through sacrificial microspheres as an in vitro model for discovering the potential effect of ovarian toxic agents. Bioact Mater 2023; 26:216-230. [PMID: 36936809 PMCID: PMC10017366 DOI: 10.1016/j.bioactmat.2023.02.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/26/2023] [Accepted: 02/26/2023] [Indexed: 03/09/2023] Open
Abstract
The bio-engineered ovary is an essential technology for treating female infertility. Especially the development of relevant in vitro models could be a critical step in a drug study. Herein, we develop a semi-opened culturing system (SOCS) strategy that maintains a 3D structure of follicles during the culture. Based on the SOCS, we further developed micro-cavity ovary (MCO) with mouse follicles by the microsphere-templated technique, where sacrificial gelatin microspheres were mixed with photo-crosslinkable gelatin methacryloyl (GelMA) to engineer a micro-cavity niche for follicle growth. The semi-opened MCO could support the follicle growing to the antral stage, secreting hormones, and ovulating cumulus-oocyte complex out of the MCO without extra manipulation. The MCO-ovulated oocyte exhibits a highly similar transcriptome to the in vivo counterpart (correlation of 0.97) and can be fertilized. Moreover, we found that a high ROS level could affect the cumulus expansion, which may result in anovulation disorder. The damage could be rescued by melatonin, but the end of cumulus expansion was 3h earlier than anticipation, validating that MCO has the potential for investigating ovarian toxic agents in vitro. We provide a novel approach for building an in vitro ovarian model to recapitulate ovarian functions and test chemical toxicity, suggesting it has the potential for clinical research in the future.
Collapse
Affiliation(s)
- Min Ye
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China
- Biomanufacturing and Engineering Living Systems, Innovation International Talents Base (111 Base), Beijing, 100084, China
| | - Yiran Shan
- MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Bingchuan Lu
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China
- Biomanufacturing and Engineering Living Systems, Innovation International Talents Base (111 Base), Beijing, 100084, China
| | - Hao Luo
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China
- Biomanufacturing and Engineering Living Systems, Innovation International Talents Base (111 Base), Beijing, 100084, China
| | - Binhan Li
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China
- Biomanufacturing and Engineering Living Systems, Innovation International Talents Base (111 Base), Beijing, 100084, China
| | - Yanmei Zhang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China
- Biomanufacturing and Engineering Living Systems, Innovation International Talents Base (111 Base), Beijing, 100084, China
| | - Zixuan Wang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China
- Biomanufacturing and Engineering Living Systems, Innovation International Talents Base (111 Base), Beijing, 100084, China
| | - Yuzhi Guo
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China
- Biomanufacturing and Engineering Living Systems, Innovation International Talents Base (111 Base), Beijing, 100084, China
| | - Liliang Ouyang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China
- Biomanufacturing and Engineering Living Systems, Innovation International Talents Base (111 Base), Beijing, 100084, China
| | - Jin Gu
- MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Zhuo Xiong
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China
- Biomanufacturing and Engineering Living Systems, Innovation International Talents Base (111 Base), Beijing, 100084, China
- Corresponding author. Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China.
| | - Ting Zhang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, China
- Biomanufacturing and Engineering Living Systems, Innovation International Talents Base (111 Base), Beijing, 100084, China
- Corresponding author. Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
20
|
Francés-Herrero E, Lopez R, Campo H, de Miguel-Gómez L, Rodríguez-Eguren A, Faus A, Pellicer A, Cervelló I. Advances of xenogeneic ovarian extracellular matrix hydrogels for in vitro follicle development and oocyte maturation. BIOMATERIALS ADVANCES 2023; 151:213480. [PMID: 37267748 DOI: 10.1016/j.bioadv.2023.213480] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 06/04/2023]
Abstract
Research aimed at preserving female fertility is increasingly using bioengineering techniques to develop new platforms capable of supporting ovarian cell function in vitro and in vivo. Natural hydrogels (alginate, collagen, and fibrin) have been the most exploited approaches; however they are biologically inert and/or biochemically simple. Thus, establishing a suitable biomimetic hydrogel from decellularized ovarian cortex (OC) extracellular matrix (OvaECM) could provide a complex native biomaterial for follicle development and oocyte maturation. The objectives of this work were (i) to establish an optimal protocol to decellularize and solubilize bovine OC, (ii) to characterize the histological, molecular, ultrastructural, and proteomic properties of the resulting tissue and hydrogel, and (iii) to assess its biocompatibility and adequacy for murine in vitro follicle growth (IVFG). Sodium dodecyl sulfate was identified as the best detergent to develop bovine OvaECM hydrogels. Hydrogels added into standard media or used as plate coatings were employed for IVFG and oocyte maturation. Follicle growth, survival, hormone production, and oocyte maturation and developmental competence were evaluated. OvaECM hydrogel-supplemented media best supported follicle survival, expansion, and hormone production, while the coatings provided more mature and competent oocytes. Overall, the findings support the xenogeneic use of OvaECM hydrogels for future human female reproductive bioengineering.
Collapse
Affiliation(s)
- Emilio Francés-Herrero
- Department of Pediatrics, Obstetrics and Gynecology, School of Medicine, University of Valencia, 46010 Valencia, Spain; IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain
| | - Rosalba Lopez
- Department of Pediatrics, Obstetrics and Gynecology, School of Medicine, University of Valencia, 46010 Valencia, Spain; IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain
| | - Hannes Campo
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Lucía de Miguel-Gómez
- Department of Pediatrics, Obstetrics and Gynecology, School of Medicine, University of Valencia, 46010 Valencia, Spain; IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain
| | - Adolfo Rodríguez-Eguren
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain
| | - Amparo Faus
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain
| | - Antonio Pellicer
- Department of Pediatrics, Obstetrics and Gynecology, School of Medicine, University of Valencia, 46010 Valencia, Spain; IVI Roma Parioli, IVI-RMA Global, 00197 Rome, Italy
| | - Irene Cervelló
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), 46026 Valencia, Spain.
| |
Collapse
|
21
|
Hu B, Wang R, Wu D, Long R, Ruan J, Jin L, Ma D, Sun C, Liao S. Prospects for fertility preservation: the ovarian organ function reconstruction techniques for oogenesis, growth and maturation in vitro. Front Physiol 2023; 14:1177443. [PMID: 37250136 PMCID: PMC10213246 DOI: 10.3389/fphys.2023.1177443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/04/2023] [Indexed: 05/31/2023] Open
Abstract
Today, fertility preservation is receiving more attention than ever. Cryopreservation, which preserves ovarian tissue to preserve fertility in young women and reduce the risk of infertility, is currently the most widely practiced. Transplantation, however, is less feasible for women with blood-borne leukemia or cancers with a high risk of ovarian metastasis because of the risk of cancer recurrence. In addition to cryopreservation and re-implantation of embryos, in vitro ovarian organ reconstruction techniques have been considered as an alternative strategy for fertility preservation. In vitro culture of oocytes in vitro Culture, female germ cells induction from pluripotent stem cells (PSC) in vitro, artificial ovary construction, and ovaria-related organoids construction have provided new solutions for fertility preservation, which will therefore maximize the potential for all patients undergoing fertility preservation. In this review, we discussed and thought about the latest ovarian organ function reconstruction techniques in vitro to provide new ideas for future ovarian disease research and fertility preservation of patients with cancer and premature ovarian failure.
Collapse
Affiliation(s)
- Bai Hu
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Renjie Wang
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Di Wu
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Long
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinghan Ruan
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Jin
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ding Ma
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chaoyang Sun
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shujie Liao
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
22
|
Zhao X, Zhang S, Gao S, Chang HM, Leung PCK, Tan J. A Novel Three-Dimensional Follicle Culture System Decreases Oxidative Stress and Promotes the Prolonged Culture of Human Granulosa Cells. ACS APPLIED MATERIALS & INTERFACES 2023; 15:15084-15095. [PMID: 36926803 PMCID: PMC10065000 DOI: 10.1021/acsami.2c18734] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 02/10/2023] [Indexed: 06/18/2023]
Abstract
Tissue engineering advancements have made it possible to modify biomaterials to reconstruct a similar three-dimensional structure of the extracellular matrix (ECM) for follicle development and to supply the required biological signals. We postulated that an artificial polysaccharide hydrogel modified with an ECM mimetic peptide may produce efficient irritation signals by binding to specific integrins providing a suitable environment for follicular development and influencing the behavior of human granulosa cells (hGCs). Laminin, an important component of the extracellular matrix, can modulate hGCs and oocyte growth. Specifically, follicles of mice were randomly divided into two-dimensional (2D) and three-dimensional (3D) culture systems established by a hydrogel modified with RGD or laminin mimetic peptides (IKVAV and YIGSR) and RGD (IYR). Our results showed that 3D cultured systems significantly improved follicle survival, growth, and viability. IYR peptides enhanced the oocyte meiosis competence. Additionally, we explored the effect of 3D culture on hGCs, which improved hGCs viability, increased the proportion of S- and G2/M-phase cells, and inhibited cell apoptosis of hGCs. On days 1 and 2, the secretion of progesterone was reduced in 3D-cultured hGCs. Notably, 3D-cultured hGCs exhibited delayed senescence, decreased oxidative stress, and elevated mitochondrial membrane potential. Moreover, the expression levels of cumulus expansion-related genes (COX2, HAS2, and PTX3) and integrin α6β1 were upregulated in 3D-cultured hGCs. In conclusion, a 3D culture utilizing hydrogels modified with Laminin-mimetic peptides can provide a durable physical environment suitable for follicular development. The laminin-mimetic peptides may regulate the biological activity of hGCs by attaching to the integrin α6β1.
Collapse
Affiliation(s)
- Xinyang Zhao
- Center
of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Tiexi District, Shenyang, Liaoning 110022, China
- Key
Laboratory of Reproductive Dysfunction Disease and Fertility Remodeling
of Liaoning Province, Shenyang, Liaoning 110022, China
| | - Siwen Zhang
- Center
of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Tiexi District, Shenyang, Liaoning 110022, China
- Key
Laboratory of Reproductive Dysfunction Disease and Fertility Remodeling
of Liaoning Province, Shenyang, Liaoning 110022, China
| | - Shan Gao
- Center
of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Tiexi District, Shenyang, Liaoning 110022, China
- Key
Laboratory of Reproductive Dysfunction Disease and Fertility Remodeling
of Liaoning Province, Shenyang, Liaoning 110022, China
| | - Hsun-Ming Chang
- Department
of Obstetrics and Gynaecology, BC Children’s Hospital Research
Institute, University of British Columbia, Vancouver, British Columbia V5Z4H4, Canada
- Reproductive
Medicine Center, Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung 404327, Taiwan
| | - Peter C. K. Leung
- Department
of Obstetrics and Gynaecology, BC Children’s Hospital Research
Institute, University of British Columbia, Vancouver, British Columbia V5Z4H4, Canada
| | - Jichun Tan
- Center
of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 39 Huaxiang Road, Tiexi District, Shenyang, Liaoning 110022, China
- Key
Laboratory of Reproductive Dysfunction Disease and Fertility Remodeling
of Liaoning Province, Shenyang, Liaoning 110022, China
| |
Collapse
|
23
|
Grosbois J, Bailie EC, Kelsey TW, Anderson RA, Telfer EE. Spatio-temporal remodelling of the composition and architecture of the human ovarian cortical extracellular matrix during in vitro culture. Hum Reprod 2023; 38:444-458. [PMID: 36721914 PMCID: PMC9977129 DOI: 10.1093/humrep/dead008] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 11/29/2022] [Indexed: 02/02/2023] Open
Abstract
STUDY QUESTION How does in vitro culture alter the human ovarian cortical extracellular matrix (ECM) network structure? SUMMARY ANSWER The ECM composition and architecture vary in the different layers of the ovarian cortex and are remodelled during in vitro culture. WHAT IS KNOWN ALREADY The ovarian ECM is the scaffold within which follicles and stromal cells are organized. Its composition and structural properties constantly evolve to accommodate follicle development and expansion. Tissue preparation for culture of primordial follicles within the native ECM involves mechanical loosening; this induces undefined modifications in the ECM network and alters cell-cell contact, leading to spontaneous follicle activation. STUDY DESIGN, SIZE, DURATION Fresh ovarian cortical biopsies were obtained from six women aged 28-38 years (mean ± SD: 32.7 ± 4.1 years) at elective caesarean section. Biopsies were cut into fragments of ∼4 × 1 × 1 mm and cultured for 0, 2, 4, or 6 days (D). PARTICIPANTS/MATERIALS, SETTING, METHODS Primordial follicle activation, stromal cell density, and ECM-related protein (collagen, elastin, fibronectin, laminin) positive area in the entire cortex were quantified at each time point using histological and immunohistological analysis. Collagen and elastin content, collagen fibre characteristics, and follicle distribution within the tissue were further quantified within each layer of the human ovarian cortex, namely the outer cortex, the mid-cortex, and the cortex-medulla junction regions. MAIN RESULTS AND THE ROLE OF CHANCE Primordial follicle activation occurred concomitantly with a loosening of the ovarian cortex during culture, characterized by an early decrease in stromal cell density from 3.6 ± 0.2 × 106 at day 0 (D0) to 2.8 ± 0.1 × 106 cells/mm3 at D2 (P = 0.033) and a dynamic remodelling of the ECM. Notably, collagen content gradually fell from 55.5 ± 1.7% positive area at D0 to 42.3 ± 1.1% at D6 (P = 0.001), while elastin increased from 1.1 ± 0.2% at D0 to 1.9 ± 0.1% at D6 (P = 0.001). Fibronectin and laminin content remained stable. Moreover, collagen and elastin distribution were uneven throughout the cortex and during culture. Analysis at the sub-region level showed that collagen deposition was maximal in the outer cortex and the lowest in the mid-cortex (69.4 ± 1.2% versus 53.8 ± 0.8% positive area, respectively, P < 0.0001), and cortical collagen staining overall decreased from D0 to D2 (65.2 ± 2.4% versus 60.6 ± 1.8%, P = 0.033) then stabilized. Elastin showed the converse distribution, being most concentrated at the cortex-medulla junction (3.7 ± 0.6% versus 0.9 ± 0.2% in the outer cortex, P < 0.0001), and cortical elastin peaked at D6 compared to D0 (3.1 ± 0.5% versus 1.3 ± 0.2%, P < 0.0001). This was corroborated by a specific signature of the collagen fibre type across the cortex, indicating a distinct phenotype of the ovarian cortical ECM depending on region and culture period that might be responsible for the spatio-temporal and developmental pattern of follicular distribution observed within the cortex. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Ovarian cortical biopsies were obtained from women undergoing caesarean sections. As such, the data obtained may not accurately reflect the ECM distribution and structure of non-pregnant women. WIDER IMPLICATIONS OF THE FINDINGS Clarifying the composition and architecture signature of the human ovarian cortical ECM provides a foundation for further exploration of ovarian microenvironments. It is also critical for understanding the ECM-follicle interactions regulating follicle quiescence and awakening, leading to improvements in both in vitro activation and in vitro growth techniques. STUDY FUNDING/COMPETING INTEREST(S) Medical Research Council grant MR/R003246/1 and Wellcome Trust Collaborative Award in Science: 215625/Z/19/Z. The authors have no conflicts to declare. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Johanne Grosbois
- Institute of Cell Biology, Hugh Robson Building, University of Edinburgh, Edinburgh, UK
| | - Emily C Bailie
- Institute of Cell Biology, Hugh Robson Building, University of Edinburgh, Edinburgh, UK
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Tom W Kelsey
- School of Computer Science, University of St Andrews, St Andrews, UK
| | - Richard A Anderson
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Evelyn E Telfer
- Institute of Cell Biology, Hugh Robson Building, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
24
|
Converse A, Zaniker EJ, Amargant F, Duncan FE. Recapitulating folliculogenesis and oogenesis outside the body: encapsulated in vitro follicle growth†. Biol Reprod 2023; 108:5-22. [PMID: 36136744 PMCID: PMC9843677 DOI: 10.1093/biolre/ioac176] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/04/2022] [Accepted: 09/21/2022] [Indexed: 01/21/2023] Open
Abstract
Folliculogenesis is a tightly coordinated process essential for generating a fertilization-competent gamete while also producing gonadal hormones that sustain endocrine function. In vitro follicle growth systems have been critical to our understanding of key events in folliculogenesis, such as gonadotropin-independent and dependent growth, steroid hormone production, and oocyte growth and maturation (cytoplasmic and meiotic). Although there are several successful follicle culture strategies, the following protocol details an encapsulated in vitro follicle growth (eIVFG) system for use with mouse ovarian follicles. Encapsulated IVFG is performed with alginate hydrogels, which are biologically inert, maintains cell-to-cell interactions between granulosa cells and the oocyte, and preserves follicle architecture as found in the ovary. The system supports follicle growth, development, and differentiation from the early primary follicle to the antral follicle stage. Moreover, post-folliculogenesis events including meiotic maturation, ovulation, and luteinization are also supported. Importantly, the culture of secondary follicles has successfully resulted in viable pups after blastocyst transfer. This alginate-based eIVFG system is versatile and has broad applications as a tool for interrogating the fundamental biology of the ovarian follicle in a controlled manner, a screening platform for toxicity and bioactivity, and a potential fertility preservation method for endangered species as well as humans.
Collapse
Affiliation(s)
- Aubrey Converse
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illnois, USA
| | - Emily J Zaniker
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illnois, USA
| | - Farners Amargant
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illnois, USA
| | - Francesca E Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illnois, USA
| |
Collapse
|
25
|
Biswas A, Ng BH, Prabhakaran VS, Chan CJ. Squeezing the eggs to grow: The mechanobiology of mammalian folliculogenesis. Front Cell Dev Biol 2022; 10:1038107. [PMID: 36531957 PMCID: PMC9756970 DOI: 10.3389/fcell.2022.1038107] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/16/2022] [Indexed: 08/25/2023] Open
Abstract
The formation of functional eggs (oocyte) in ovarian follicles is arguably one of the most important events in early mammalian development since the oocytes provide the bulk genetic and cytoplasmic materials for successful reproduction. While past studies have identified many genes that are critical to normal ovarian development and function, recent studies have highlighted the role of mechanical force in shaping folliculogenesis. In this review, we discuss the underlying mechanobiological principles and the force-generating cellular structures and extracellular matrix that control the various stages of follicle development. We also highlight emerging techniques that allow for the quantification of mechanical interactions and follicular dynamics during development, and propose new directions for future studies in the field. We hope this review will provide a timely and useful framework for future understanding of mechano-signalling pathways in reproductive biology and diseases.
Collapse
Affiliation(s)
- Arikta Biswas
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Boon Heng Ng
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | | | - Chii Jou Chan
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| |
Collapse
|
26
|
Ghorbani S, Eyni H, Norahan MH, Zarrintaj P, Urban N, Mohammadzadeh A, Mostafavi E, Sutherland DS. Advanced bioengineering of female germ cells to preserve fertility. Biol Reprod 2022; 107:1177-1204. [PMID: 35947985 PMCID: PMC10144627 DOI: 10.1093/biolre/ioac160] [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: 05/06/2022] [Revised: 07/29/2022] [Accepted: 08/02/2022] [Indexed: 11/14/2022] Open
Abstract
Oogenesis and folliculogenesis are considered as complex and species-specific cellular differentiation processes, which depend on the in vivo ovarian follicular environment and endocrine cues. Considerable efforts have been devoted to driving the differentiation of female primordial germ cells toward mature oocytes outside of the body. The recent experimental attempts have laid stress on offering a suitable microenvironment to assist the in vitro folliculogenesis and oogenesis. Despite developing a variety of bioengineering techniques and generating functional mature gametes through in vitro oogenesis in earlier studies, we still lack knowledge of appropriate microenvironment conditions for building biomimetic culture systems for female fertility preservation. Therefore, this review paper can provide a source for a large body of scientists developing cutting-edge in vitro culture systems for female germ cells or setting up the next generation of reproductive medicine as feasible options for female infertility treatment. The focal point of this review outlines advanced bioengineering technologies such as 3D biofabricated hydrogels/scaffolds and microfluidic systems utilized with female germlines for fertility preservation through in vitro folliculogenesis and oogenesis.
Collapse
Affiliation(s)
- Sadegh Ghorbani
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus, Denmark
| | - Hossein Eyni
- Cellular and Molecular Research Center, School of Medicine, Iran University of Medical Science, Tehran, Iran
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Mohammad Hadi Norahan
- School of Engineering and Sciences, Tecnologico de Monterrey Unviersity, Monterrey, NL, Mexico
| | - Payam Zarrintaj
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, USA
| | - Nadine Urban
- Freiburg Centre for Interactive Materials and Bioinspired Technology, University of Freiburg, Freiburg, Germany
| | | | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Duncan S Sutherland
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus, Denmark
| |
Collapse
|
27
|
Di Berardino C, Peserico A, Capacchietti G, Zappacosta A, Bernabò N, Russo V, Mauro A, El Khatib M, Gonnella F, Konstantinidou F, Stuppia L, Gatta V, Barboni B. High-Fat Diet and Female Fertility across Lifespan: A Comparative Lesson from Mammal Models. Nutrients 2022; 14:nu14204341. [PMID: 36297035 PMCID: PMC9610022 DOI: 10.3390/nu14204341] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/10/2022] [Accepted: 10/14/2022] [Indexed: 11/18/2022] Open
Abstract
Female reproduction focuses mainly on achieving fully grown follicles and competent oocytes to be successfully fertilized, as well as on nourishing the developing offspring once pregnancy occurs. Current evidence demonstrates that obesity and/or high-fat diet regimes can perturbate these processes, leading to female infertility and transgenerational disorders. Since the mechanisms and reproductive processes involved are not yet fully clarified, the present review is designed as a systematic and comparative survey of the available literature. The available data demonstrate the adverse influences of obesity on diverse reproductive processes, such as folliculogenesis, oogenesis, and embryo development/implant. The negative reproductive impact may be attributed to a direct action on reproductive somatic and germinal compartments and/or to an indirect influence mediated by the endocrine, metabolic, and immune axis control systems. Overall, the present review highlights the fragmentation of the current information limiting the comprehension of the reproductive impact of a high-fat diet. Based on the incidence and prevalence of obesity in the Western countries, this topic becomes a research challenge to increase self-awareness of dietary reproductive risk to propose solid and rigorous preventive dietary regimes, as well as to develop targeted pharmacological interventions.
Collapse
Affiliation(s)
- Chiara Di Berardino
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Alessia Peserico
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
- Correspondence:
| | - Giulia Capacchietti
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Alex Zappacosta
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Nicola Bernabò
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council, A. Buzzati-Traverso Campus, via E. Ramarini 32, Monterotondo Scalo, 00015 Rome, Italy
| | - Valentina Russo
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Annunziata Mauro
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Mohammad El Khatib
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Francesca Gonnella
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Fani Konstantinidou
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Liborio Stuppia
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Valentina Gatta
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Barbara Barboni
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| |
Collapse
|
28
|
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: 6] [Impact Index Per Article: 2.0] [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
|
29
|
Babayev E, Xu M, Shea LD, Woodruff TK, Duncan FE. Follicle isolation methods reveal plasticity of granulosa cell steroidogenic capacity during mouse in vitro follicle growth. Mol Hum Reprod 2022; 28:6693628. [PMID: 36069625 PMCID: PMC9802420 DOI: 10.1093/molehr/gaac033] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 07/26/2022] [Indexed: 01/07/2023] Open
Abstract
Follicles are the functional unit of the ovary and several methods have been developed to grow follicles ex vivo, which recapitulate key events of oogenesis and folliculogenesis. Enzymatic digestion protocols are often used to increase the yield of follicles from the ovary. However, the impact of these protocols on the outermost theca and granulosa cells, and thereby follicle function, is not well defined. To investigate the impact of enzymatic digestion on follicle function, we collected preantral follicles from CD1 mice either by enzymatic digestion (Enzy-FL) or mechanical isolation (Mech-FL) and compared follicle growth, steroidogenesis and cell differentiation within an encapsulated in vitro follicle growth system which maintains the 3D architecture of the oocyte and its surrounding somatic cells. Follicles were encapsulated in 0.5% alginate and cultured for 8 days. Compared with Enzy-FL, Mech-FL grew more rapidly and produced significantly higher levels of androstenedione, estradiol and progesterone. The expression of theca-interstitial cell marker genes, Cyp17a1, which encodes 17-hydroxylase/17, 20-lyase and catalyzes the hydroxylation of pregnenolone and progesterone to 17-hydroxypregnenolone and 17-hydroxyprogesterone, and the conversion of these products into dehydroepiandrosterone and androstenedione, and Star, which encodes a transport protein essential for cholesterol entry into mitochondria, were also higher in Mech-FL than in Enzy-FL. Mech-FL maintained an intact theca-interstitial layer on the outer edge of the follicle that phenocopied in vivo patterns as confirmed by alkaline phosphatase staining, whereas theca-interstitial cells were absent from Enzy-FL from the onset of culture. Therefore, preservation of the theca cell layer at the onset of culture better supports follicle growth and function. Interestingly, granulosa cells in the outermost layers of Enzy-FL expressed CYP17A1 by Day 4 of culture while maintaining inhibin α-subunit expression and a cuboidal nucleus. Thus, in the absence of theca-interstitial cells, granulosa cells have the potential to differentiate into androgen-producing cells. This work may have implications for human follicle culture, where enzymatic isolation is required owing to the density of the ovarian cortex.
Collapse
Affiliation(s)
| | | | - Lonnie D Shea
- Member of the Oncofertility Consortium, Michigan State University, East Lansing, MI, USA,Institute of Bionanotechnology in Medicine, Northwestern University, Chicago, IL, USA,Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL, USA
| | - Teresa K Woodruff
- Correspondence address. Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, 303 E. Superior Street, Lurie 10-109, Chicago, IL 60611, USA. E-mail: (F.E.D.); Department of Obstetrics and Gynecology and Department of Biomedical Engineering, Michigan State University, 965 Wilson Road, Room A626B, East Lansing, MI 48824-1316, USA. E-mail: (T.K.W.)
| | - Francesca E Duncan
- Correspondence address. Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, 303 E. Superior Street, Lurie 10-109, Chicago, IL 60611, USA. E-mail: (F.E.D.); Department of Obstetrics and Gynecology and Department of Biomedical Engineering, Michigan State University, 965 Wilson Road, Room A626B, East Lansing, MI 48824-1316, USA. E-mail: (T.K.W.)
| |
Collapse
|
30
|
Zhang D, Ding C, Duan T, Zhou Q. Applications of Hydrogels in Premature Ovarian Failure and Intrauterine Adhesion. FRONTIERS IN MATERIALS 2022; 9. [DOI: 10.3389/fmats.2022.942957] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Premature ovarian failure (POF) and intrauterine adhesion (IUA) that easily lead to reduced fertility in premenopausal women are two difficult diseases to treat in obstetrics and gynecology. Hormone therapy, in vitro fertilization and surgical treatments do not completely restore fertility. The advent of hydrogels offers new hope for the treatment of POF and IUA. Hydrogels are noncytotoxic and biodegradable, and do not cause immune rejection or inflammatory reactions. Drug delivery and stem cell delivery are the main application forms. Hydrogels are a local drug delivery reservoir, and the control of drug release is achieved by changing the physicochemical properties. The porous properties and stable three-dimensional structure of hydrogels support stem cell growth and functions. In addition, hydrogels are promising biomaterials for increasing the success rate of ovarian tissue transplantation. Hydrogel-based in vitro three-dimensional culture of follicles drives the development of artificial ovaries. Hydrogels form a barrier at the site of injury and have antibacterial, antiadhesive and antistenosis properties for IUA treatment. In this review, we evaluate the physicochemical properties of hydrogels, and focus on the latest applications of hydrogels in POF and IUA. We also found the limitations on clinical application of hydrogel and provide future prospects. Artificial ovary as the future of hydrogel in POF is worth studying, and 3D bioprinting may help the mass production of hydrogels.
Collapse
|
31
|
Herta AC, Mengden L, Akin N, Billooye K, Coucke W, Leersum J, Cava-Cami B, Saucedo-Cuevas L, Klamt F, Smitz J, Anckaert E. Characterization of carbohydrate metabolism in in vivo and in vitro grown and matured mouse antral follicles. Biol Reprod 2022; 107:998-1013. [PMID: 35717588 DOI: 10.1093/biolre/ioac124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 02/14/2022] [Accepted: 06/12/2022] [Indexed: 11/13/2022] Open
Abstract
Establishing an ideal human follicle culture system for oncofertility patients relies mainly on animal models since donor tissue is scarce and often of suboptimal quality. The in vitro system developed in our laboratory supports the growth of prepubertal mouse secondary follicles up to mature oocytes. Given the importance of glucose in preparing the oocyte for proper maturation, a baseline characterization of follicle metabolism both in the culture system and in vivo was carried out. Markers of glucose-related pathways (glycolysis, tricarboxylic acid (TCA) cycle, pentose phosphate pathway (PPP), polyol pathway, hexosamine biosynthesis pathway (HBP)) as well as for the antioxidant capacity were measured in the different follicle cell types by both enzymatic activities (spectrophotometric detection) and gene expression (qPCR). This study confirmed that in vivo the somatic cells, mainly granulosa, exhibit intense glycolytic activity, while oocytes perform PPP. Throughout the final maturation step, oocytes in vivo and in vitro showed steady levels for all the key enzymes and metabolites. On the other hand, ovulation triggers a boost of pyruvate and lactate uptake in cumulus cells in vivo, consumes reduced nicotinamide adenine dinucleotide phosphate (NADPH) and increases TCA cycle and small molecules antioxidant capacity (SMAC) activities, while in vitro, the metabolic upregulation in all the studied pathways is limited. This altered metabolic pattern might be a consequence of cell exhaustion because of culture conditions, impeding cumulus cells to fulfil their role in providing proper support for acquiring oocyte competence. SUMMARY SENTENCE: In vitro cultured mouse follicles exhibit altered glycolytic activity and redox metabolism in the somatic compartment during meiotic maturation.
Collapse
Affiliation(s)
- Anamaria-Cristina Herta
- Follicle Biology Laboratory (FOBI), Vrije Universiteit Brussel (VUB), Brussels, 1090, Belgium
| | - Lucia Mengden
- Laboratory of Cellular Biochemistry, Department of Biochemistry, ICBS, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre (RS), 90035003, Brazil
| | - Nazli Akin
- Follicle Biology Laboratory (FOBI), Vrije Universiteit Brussel (VUB), Brussels, 1090, Belgium
| | - Katy Billooye
- Follicle Biology Laboratory (FOBI), Vrije Universiteit Brussel (VUB), Brussels, 1090, Belgium
| | - Wim Coucke
- Freelance statistician, Brugstraat 107, 3001 Heverlee, Belgium
| | - Julia Leersum
- Follicle Biology Laboratory (FOBI), Vrije Universiteit Brussel (VUB), Brussels, 1090, Belgium
| | - Berta Cava-Cami
- Follicle Biology Laboratory (FOBI), Vrije Universiteit Brussel (VUB), Brussels, 1090, Belgium
| | - Laura Saucedo-Cuevas
- Follicle Biology Laboratory (FOBI), Vrije Universiteit Brussel (VUB), Brussels, 1090, Belgium
| | - Fábio Klamt
- Laboratory of Cellular Biochemistry, Department of Biochemistry, ICBS, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre (RS), 90035003, Brazil
| | - Johan Smitz
- Follicle Biology Laboratory (FOBI), Vrije Universiteit Brussel (VUB), Brussels, 1090, Belgium
| | - Ellen Anckaert
- Follicle Biology Laboratory (FOBI), Vrije Universiteit Brussel (VUB), Brussels, 1090, Belgium
| |
Collapse
|
32
|
Umehara T, Winstanley YE, Andreas E, Morimoto A, Williams EJ, Smith KM, Carroll J, Febbraio MA, Shimada M, Russell DL, Robker RL. Female reproductive life span is extended by targeted removal of fibrotic collagen from the mouse ovary. SCIENCE ADVANCES 2022; 8:eabn4564. [PMID: 35714185 PMCID: PMC9205599 DOI: 10.1126/sciadv.abn4564] [Citation(s) in RCA: 106] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
The female ovary contains a finite number of oocytes, and their release at ovulation becomes sporadic and disordered with aging and with obesity, leading to loss of fertility. Understanding the molecular defects underpinning this pathology is essential as age of childbearing and obesity rates increase globally. We identify that fibrosis within the ovarian stromal compartment is an underlying mechanism responsible for impaired oocyte release, which is initiated by mitochondrial dysfunction leading to diminished bioenergetics, oxidative damage, inflammation, and collagen deposition. Furthermore, antifibrosis drugs (pirfenidone and BGP-15) eliminate fibrotic collagen and restore ovulation in reproductively old and obese mice, in association with dampened M2 macrophage polarization and up-regulated MMP13 protease. This is the first evidence that ovarian fibrosis is reversible and indicates that drugs targeting mitochondrial metabolism may be a viable therapeutic strategy for women with metabolic disorders or advancing age to maintain ovarian function and extend fertility.
Collapse
Affiliation(s)
- Takashi Umehara
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - Yasmyn E. Winstanley
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Eryk Andreas
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Atsushi Morimoto
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Elisha J. Williams
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Kirsten M. Smith
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - John Carroll
- Development and Stem Cells Program and Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Mark A. Febbraio
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Masayuki Shimada
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - Darryl L. Russell
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Rebecca L. Robker
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
- Development and Stem Cells Program and Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Corresponding author.
| |
Collapse
|
33
|
Zivari-Ghader T, Dolati S, Mehdizadeh A, Davaran S, Rashidi MR, Yousefi M. Recent scaffold-based tissue engineering approaches in premature ovarian failure treatment. J Tissue Eng Regen Med 2022; 16:605-620. [PMID: 35511799 DOI: 10.1002/term.3306] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/09/2022] [Accepted: 04/11/2022] [Indexed: 11/10/2022]
Abstract
Recently, tissue engineering and regenerative medicine have received significant attention with outstanding advances. The main scope of this technology is to recover the damaged tissues and organs or to maintain and improve their function. One of the essential fields in tissue engineering is scaffold designing and construction, playing an integral role in damaged tissues reconstruction and repair. However, premature ovarian failure (POF) is a disorder causing many medical and psychological problems in women. POF treatment using tissue engineering and various scaffold has recently made tremendous and promising progress. Due to the importance of the subject, we have summarized the recently examined scaffolds in the treatment of POF in this review.
Collapse
Affiliation(s)
- Tayyebeh Zivari-Ghader
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sanam Dolati
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soodabeh Davaran
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Rashidi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
34
|
Haghshenas M, Tavana S, Zand E, Montazeri L, Fathi R. Mouse ovarian follicle growth in an amniotic membrane-based hydrogel. J Biomater Appl 2022; 37:563-574. [PMID: 35451867 DOI: 10.1177/08853282221094193] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Three-dimensional cultures of follicles on ECM-based scaffolds can be an approach for women who become infertile after cancer treatments. Human amniotic membrane (HAM) is extensively employed in tissue engineering because of its unique properties. We cultured mouse pre-antral follicles in a hydrogel derived from decellularized amniotic membrane (DAM) combined with alginate (ALG) to improve ovarian follicle culture. HAM was decellularized. Quantitative (nuclear contents, collagen, glycosaminoglycan [GAG]) and qualitative (DAPI, H&E, Masson's trichrome, Alcian blue, scanning electron microscopy assessments were performed. Then, we created an amniotic membrane-based hydrogel (AMBH) and conducted AMBH characterization assays (rheology, MTS, degradation rate). Isolated mouse pre-antral follicles were cultured in 15 mg/mL AMBH (AMBH15), 30 mg/mL AMBH (AMBH30), or 45 mg/mL AMBH (AMBH45). ALG hydrogel was the control group. Follicular diameters, estradiol hormone secretion rate, follicular morphology, and the follicle antral and degeneration rate were examined. Quantitative and qualitative assays indicated successful decellularization. AMBH characterization assays showed that the ALG hydrogel had more appropriate gelation and slower degradation than AMBH. There was a statistically higher antral follicle formation rate in the AMBH45 group (p < .05) compared to the AMBH30 and AMBH15 groups and less (p < .05) degenerated follicles. There was no significant difference with the ALG group. Diameter and estradiol hormone secretion in the AMBH45 group were not significantly higher than the ALG group. Although decellularization was confirmed and the viscoelastic parameters of AMBH support follicle culture, there was no significant effect on ovarian follicle maturation compared to the ALG control group.
Collapse
Affiliation(s)
- Mohammad Haghshenas
- 438298Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.,48499Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Somayeh Tavana
- 438298Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Elnaz Zand
- 438298Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Leila Montazeri
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Rouhollah Fathi
- 438298Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| |
Collapse
|
35
|
Matsushige C, Xu X, Miyagi M, Zuo YY, Yamazaki Y. RGD-modified dextran hydrogel promotes follicle growth in three-dimensional ovarian tissue culture in mice. Theriogenology 2022; 183:120-131. [PMID: 35247849 PMCID: PMC9005264 DOI: 10.1016/j.theriogenology.2022.02.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/06/2022] [Accepted: 02/07/2022] [Indexed: 10/19/2022]
Abstract
In vitro follicle growth is a promising technology to preserve fertility for cancer patients. We previously developed a three-dimensional (3-D) ovarian tissue culture system supported by mouse tumor cell-derived Matrigel. When murine ovarian tissues at 14 days old were cultured in Matrigel drops, antrum formation and oocyte competence were significantly enhanced compared with those cultured without Matrigel. In this study, we tested whether nonanimal-derived dextran hydrogels can support a 3-D ovarian tissue culture. We employed chemically defined dextran hydrogels consisting of dextran polymers crosslinked with polyethylene glycol (PEG)-based cell-degradable crosslinker. To determine the optimal gel elasticity for the 3-D tissue culture, we measured Young's modulus of dextran hydrogels at four concentrations (1.75, 2.25, 2.75, and 3.25 mmol/L), and cultured ovarian tissues in these gels for 7 days. As a result, 2.25 mmol/L dextran hydrogel with Young's modulus of 224 Pa was appropriate to provide physical support as well as to promote follicle expansion in the 3-D system. To mimic the natural extracellular matrix (ECM) environment, we modified the dextran hydrogels with two bioactive factors: ECM-derived Arg-Gly-Asp (RGD) peptides as a cell-adhesive factor, and activin A. The ovarian tissues were cultured in 2.25 mmol/L dextran hydrogels under four different conditions: Activin-/RGD- (A-R-), A + R-, A-R+, and A + R+. On Day 7 of culture, follicle and oocyte sizes were significantly increased in the RGD-modified conditions compared with those without RGD. The RGD-modified hydrogels also promoted mRNA levels of steroidogenic-related genes and estradiol production in the 3-D ovarian tissue culture. In vitro maturation and developmental competence of follicular oocytes were remarkably improved in the presence of RGD. In particular, blastocyst embryos were obtained only from A-R+ or A+R+ conditions after in vitro fertilization. We also determined synergistic effects of the RGD peptides and activin A on follicle growth and oocyte development in the 3-D tissue culture. In conclusion, our results suggest that RGD-modified dextran hydrogels provide an ECM-mimetic bioactive environment to support folliculogenesis in a 3-D ovarian tissue culture system.
Collapse
|
36
|
Liao B, Qi X, Yun C, Qiao J, Pang Y. Effects of Androgen Excess-Related Metabolic Disturbances on Granulosa Cell Function and Follicular Development. Front Endocrinol (Lausanne) 2022; 13:815968. [PMID: 35237237 PMCID: PMC8883052 DOI: 10.3389/fendo.2022.815968] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/06/2022] [Indexed: 01/24/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common reproductive endocrine disease in women of reproductive age. Ovarian dysfunction including abnormal steroid hormone synthesis and follicular arrest play a vital role in PCOS pathogenesis. Hyperandrogenemia is one of the important characteristics of PCOS. However, the mechanism of regulation and interaction between hyperandrogenism and ovulation abnormalities are not clear. To investigate androgen-related metabolic state in granulosa cells of PCOS patients, we identified the transcriptome characteristics of PCOS granulosa cells by RNA-seq. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis of differentially expressed genes (DEGs) revealed that genes enriched in lipid metabolism pathway, fatty acid biosynthetic process and ovarian steroidogenesis pathway were abnormally expressed in PCOS granulosa cells in comparison with that in control. There are close interactions among these three pathways as identified by analysis of the protein-protein interaction (PPI) network of DEGs. Furthermore, in vitro mouse follicle culture system was established to explore the effect of high androgen and its related metabolic dysfunction on follicular growth and ovulation. RT-qPCR results showed that follicles cultured with dehydroepiandrosterone (DHEA) exhibited decreased expression levels of cumulus expansion-related genes (Has2, Ptx3, Tnfaip6 and Adamts1) and oocyte maturation-related genes (Gdf9 and Bmp15), which may be caused by impaired steroid hormone synthesis and lipid metabolism, thus inhibited follicular development and ovulation. Furthermore, the inhibition effect of DHEA on follicle development and ovulation was ameliorated by flutamide, an androgen receptor (AR) antagonist, suggesting the involvement of AR signaling. In summary, our study offers new insights into understanding the role of androgen excess induced granulosa cell metabolic disorder in ovarian dysfunction of PCOS patients.
Collapse
Affiliation(s)
- Baoying Liao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing, China
| | - Xinyu Qi
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing, China
| | - Chuyu Yun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Science, Peking University, Ministry of Education, Beijing, China
| | - Jie Qiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing, China
| | - Yanli Pang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing, China
- *Correspondence: Yanli Pang,
| |
Collapse
|
37
|
Xu J, Zelinski MB. Oocyte quality following in vitro follicle development†. Biol Reprod 2021; 106:291-315. [PMID: 34962509 PMCID: PMC9004734 DOI: 10.1093/biolre/ioab242] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/15/2021] [Accepted: 12/24/2021] [Indexed: 12/30/2022] Open
Abstract
In vitro follicle development (IVFD) is an adequate model to obtain basic knowledge of folliculogenesis and provides a tool for ovarian toxicity screening. IVFD yielding competent oocytes may also offer an option for fertility and species preservation. To promote follicle growth and oocyte maturation in vitro, various culture systems are utilized for IVFD in rodents, domestic animals, wild animals, nonhuman primates, and humans. Follicle culture conditions have been improved by optimizing gonadotropin levels, regulatory factors, nutrient supplements, oxygen concentration, and culture matrices. This review summarizes quality assessment of oocytes generated from in vitro-developed antral follicles from the preantral stage, including oocyte epigenetic and genetic profile, cytoplasmic and nuclear maturation, preimplantation embryonic development following in vitro fertilization, as well as pregnancy and live offspring after embryo transfer. The limitations of oocyte quality evaluation following IVFD and the gaps in our knowledge of IVFD to support proper oocyte development are also discussed. The information may advance our understanding of the requirements for IVFD, with a goal of producing competent oocytes with genetic integrity to sustain embryonic development resulting in healthy offspring.
Collapse
Affiliation(s)
- Jing Xu
- Correspondence: Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR 97006, USA. Tel: +1 5033465411; Fax: +1 5033465585; E-mail:
| | - Mary B Zelinski
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA,Department of Obstetrics and Gynecology, School of Medicine, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
38
|
Bernabò N, Di Berardino C, Capacchietti G, Peserico A, Buoncuore G, Tosi U, Crociati M, Monaci M, Barboni B. In Vitro Folliculogenesis in Mammalian Models: A Computational Biology Study. Front Mol Biosci 2021; 8:737912. [PMID: 34859047 PMCID: PMC8630647 DOI: 10.3389/fmolb.2021.737912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 10/04/2021] [Indexed: 11/27/2022] Open
Abstract
In vitro folliculogenesis (ivF) has been proposed as an emerging technology to support follicle growth and oocyte development. It holds a great deal of attraction from preserving human fertility to improving animal reproductive biotechnology. Despite the mice model, where live offspring have been achieved,in medium-sized mammals, ivF has not been validated yet. Thus, the employment of a network theory approach has been proposed for interpreting the large amount of ivF information collected to date in different mammalian models in order to identify the controllers of the in vitro system. The WoS-derived data generated a scale-free network, easily navigable including 641 nodes and 2089 links. A limited number of controllers (7.2%) are responsible for network robustness by preserving it against random damage. The network nodes were stratified in a coherent biological manner on three layers: the input was composed of systemic hormones and somatic-oocyte paracrine factors; the intermediate one recognized mainly key signaling molecules such as PI3K, KL, JAK-STAT, SMAD4, and cAMP; and the output layer molecules were related to functional ivF endpoints such as the FSH receptor and steroidogenesis. Notably, the phenotypes of knock-out mice previously developed for hub.BN indirectly corroborate their biological relevance in early folliculogenesis. Finally, taking advantage of the STRING analysis approach, further controllers belonging to the metabolic axis backbone were identified, such as mTOR/FOXO, FOXO3/SIRT1, and VEGF, which have been poorly considered in ivF to date. Overall, this in silico study identifies new metabolic sensor molecules controlling ivF serving as a basis for designing innovative diagnostic and treatment methods to preserve female fertility.
Collapse
Affiliation(s)
- Nicola Bernabò
- Unit of Basic and Applied Biosciences, University of Teramo, Teramo, Italy
- National Research Council, Institute of Biochemistry and Cell Biology, Rome, Italy
| | | | | | - Alessia Peserico
- Unit of Basic and Applied Biosciences, University of Teramo, Teramo, Italy
| | - Giorgia Buoncuore
- Unit of Basic and Applied Biosciences, University of Teramo, Teramo, Italy
| | - Umberto Tosi
- Unit of Basic and Applied Biosciences, University of Teramo, Teramo, Italy
| | - Martina Crociati
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
- Centre for Perinatal and Reproductive Medicine, University of Perugia, Perugia, Italy
| | - Maurizio Monaci
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
- Centre for Perinatal and Reproductive Medicine, University of Perugia, Perugia, Italy
| | - Barbara Barboni
- Unit of Basic and Applied Biosciences, University of Teramo, Teramo, Italy
| |
Collapse
|
39
|
Pais AS, Reis S, Laranjo M, Caramelo F, Silva F, Botelho MF, Almeida-Santos T. The challenge of ovarian tissue culture: 2D versus 3D culture. J Ovarian Res 2021; 14:147. [PMID: 34724957 PMCID: PMC8561954 DOI: 10.1186/s13048-021-00892-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 10/06/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Cryopreservation of ovarian tissue is a powerful technique for preserving female fertility, as it can restore fertility and endocrine function. To increase the longevity of the transplant and decrease the risk of reimplantation of neoplastic cells, several studies have been carried out with culture of ovarian tissue. The aim of this study was to compare a conventional (2D) culture with an alginate matrix three-dimensional (3D) model for ovarian tissue culture. RESULTS The ovarian tissue culture within the alginate matrix (3D) was similar to 2D culture, regarding follicular density and cell apoptosis in follicles and stroma. The proliferation rate remained stable in both models for follicles, but for stromal cell proliferation it decreased only in 3D culture (p = 0.001). At 24 h of culture, cytotoxicity was lower in the 3D model (p = 0.006). As culture time increased, cytotoxicity seemed similar. Degradation of the tissue was suggested by the histological score analysis of tissue morphology after 72 h of culture. Tissue injury was greater (p = 0.01) in 3D culture due to higher interstitial oedema (p = 0.017) and tissue necrosis (p = 0.035). CONCLUSION According to our results, 3D culture of ovarian tissue has no advantage over 2Dculture; it is more time consuming and difficult to perform and has worse reproducibility.
Collapse
Affiliation(s)
- Ana Sofia Pais
- Reproductive Medicine Unit, Centro Hospitalar e Universitário de Coimbra (CHUC), E.P.E., Coimbra, Portugal.
- Obstetrics Department, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Biophysics Institute of Faculty of Medicine, Coimbra, Portugal.
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal.
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal.
| | - Sandra Reis
- Reproductive Medicine Unit, Centro Hospitalar e Universitário de Coimbra (CHUC), E.P.E., Coimbra, Portugal
- Center for Neuroscience and Cell Biology (CNC), CIBB, Azinhaga de Santa Comba, Celas, University of Coimbra, Coimbra, Portugal
- PhD Programme in Experimental Biology and Biomedicine, IIIUC - Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Mafalda Laranjo
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Biophysics Institute of Faculty of Medicine, Coimbra, Portugal
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Francisco Caramelo
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Biophysics Institute of Faculty of Medicine, Coimbra, Portugal
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Fátima Silva
- Pathology Unit, Centro Hospitalar e Universitário de Coimbra (CHUC), E.P.E., Coimbra, Portugal
| | - Maria Filomena Botelho
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR) Area of Environment Genetics and Oncobiology (CIMAGO), Biophysics Institute of Faculty of Medicine, Coimbra, Portugal
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Teresa Almeida-Santos
- Reproductive Medicine Unit, Centro Hospitalar e Universitário de Coimbra (CHUC), E.P.E., Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- Center for Neuroscience and Cell Biology (CNC), CIBB, Azinhaga de Santa Comba, Celas, University of Coimbra, Coimbra, Portugal
- University of Coimbra, Faculty of Medicine, Coimbra, Portugal
| |
Collapse
|
40
|
Mechanical mapping of mammalian follicle development using Brillouin microscopy. Commun Biol 2021; 4:1133. [PMID: 34580426 PMCID: PMC8476509 DOI: 10.1038/s42003-021-02662-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 09/08/2021] [Indexed: 02/08/2023] Open
Abstract
In early mammalian development, the maturation of follicles containing the immature oocytes is an important biological process as the functional oocytes provide the bulk genetic and cytoplasmic materials for successful reproduction. Despite recent work demonstrating the regulatory role of mechanical stress in oocyte growth, quantitative studies of ovarian mechanical properties remain lacking both in vivo and ex vivo. In this work, we quantify the material properties of ooplasm, follicles and connective tissues in intact mouse ovaries at distinct stages of follicle development using Brillouin microscopy, a non-invasive tool to probe mechanics in three-dimensional (3D) tissues. We find that the ovarian cortex and its interior stroma have distinct material properties associated with extracellular matrix deposition, and that intra-follicular mechanical compartments emerge during follicle maturation. Our work provides an alternative approach to study the role of mechanics in follicle morphogenesis and might pave the way for future understanding of mechanotransduction in reproductive biology, with potential implications for infertility diagnosis and treatment.
Collapse
|
41
|
Tomaszewski CE, DiLillo KM, Baker BM, Arnold KB, Shikanov A. Sequestered cell-secreted extracellular matrix proteins improve murine folliculogenesis and oocyte maturation for fertility preservation. Acta Biomater 2021; 132:313-324. [PMID: 33766798 DOI: 10.1016/j.actbio.2021.03.041] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 03/14/2021] [Accepted: 03/16/2021] [Indexed: 12/16/2022]
Abstract
Synthetic matrices offer a high degree of control and tunability for mimicking extracellular matrix functions of native tissue, allowing the study of disease and development in vitro. In this study, we functionalized degradable poly(ethylene glycol) hydrogels with extracellular matrix (ECM)-sequestering peptides aiming to recapitulate the native ECM composition for culture and maturation of ovarian follicular organoids. We hypothesized that ECM-sequestering peptides would facilitate deposition and retention of cell-secreted ECM molecules, thereby recreating cell-matrix interactions in otherwise bioinert PEG hydrogels. Specifically, heparin-binding peptide from antithrombin III (HBP), heparan sulfate binding peptide derived from laminin (AG73), basement membrane binder peptide (BMB), and heparan sulfate binding region of placental growth factor 2 (RRR) tethered to a PEG hydrogel significantly improved follicle survival, growth and maturation compared to PEG-Cys, a mechanically similar but biologically inert control. Immunohistochemical analysis of the hydrogel surrounding cultured follicles confirmed sequestration and retention of laminin, collagen I, perlecan, and fibronectin in ECM-sequestering hydrogels but not in bioinert PEG-Cys hydrogels. The media from follicles cultured in PEG-AG73, PEG-BMB, and PEG-RRR also had significantly higher concentrations of factors known to regulate follicle development compared to PEG-Cys. PEG-AG73 and PEG-BMB were the most beneficial for promoting follicle maturation, likely because AG73 and BMB mimic basement membrane interactions which are crucial for follicle development. Here we have shown that functionalizing PEG with ECM-sequestering peptides allows cell-secreted ECM to be retained within the hydrogels, restoring critical cell-matrix interactions and promoting healthy organoid development in a fully synthetic culture system. STATEMENT OF SIGNIFICANCE: Here we present a novel approach for sequestering and retaining cell-secreted extracellular matrix in a fully synthetic material for organoid culture. We have engineered a biomimetic poly(ethylene glycol) hydrogel functionalized with extracellular matrix-binding peptides to recapitulate the ovarian microenvironment. Incorporation of these peptides allows ovarian follicles to recreate their native matrix with the sequestered ECM that subsequently binds growth factors, facilitating follicle maturation. The novel design resulted in improved outcomes of folliculogenesis, potentially developing a fertility preservation option for young women undergoing sterilizing treatments for cancer. The fully synthetic and modular nature of this biomimetic material holds promise for other tissue engineering applications as it allows encapsulated cells to rebuild their native microenvironments in vitro.
Collapse
Affiliation(s)
- Claire E Tomaszewski
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Katarina M DiLillo
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Brendon M Baker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Kelly B Arnold
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Ariella Shikanov
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, 48109, USA; Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, 48109, USA; Macromolecular Science and Engineering, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
42
|
Xiang D, Liu Y, Zhou E, Wang Y. Advances in the applications of polymer biomaterials for in vitro follicle culture. Biomed Pharmacother 2021; 140:111422. [PMID: 34098195 DOI: 10.1016/j.biopha.2021.111422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 02/05/2021] [Accepted: 02/16/2021] [Indexed: 12/17/2022] Open
Abstract
The ovarian reserve (OR) indicates ovarian function by representing the quantity and quality of ovarian follicles, and it gradually decreases with increasing age. With the prolongation of women's lives, the protection provided by estrogen is lost for decades in postmenopausal women, and the related cardiovascular and cerebrovascular diseases, osteoporosis, and decreased immunity are the main risk factors affecting women's quality of life and longevity. Pharmacologic hormone replacement therapy (PHRT) has been controversial, and the construction of artificial ovary (AO) has attracted increasing attention. The most critical step of AO generation is the establishment of an in vitro culture (IVC) system to support the development of isolated follicles. This article mainly compares the advantages and disadvantages of different polymer biomaterials for use in follicle IVC, provides theoretical support for the development and construction of the follicle IVC system using natural biological materials, and provides a theoretical basis for establishing mature AO technology.
Collapse
Affiliation(s)
- Du Xiang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, 169 Donghu Road, Wuhan, Hubei 430071, China
| | - Yang Liu
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, 169 Donghu Road, Wuhan, Hubei 430071, China
| | - Encheng Zhou
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, 169 Donghu Road, Wuhan, Hubei 430071, China
| | - Yanfeng Wang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, 169 Donghu Road, Wuhan, Hubei 430071, China.
| |
Collapse
|
43
|
Jiang K, Zhang J, Huang Y, Wang Y, Xiao S, Hadden MK, Woodruff TK, Sun J. A platform utilizing Drosophila ovulation for nonhormonal contraceptive screening. Proc Natl Acad Sci U S A 2021; 118:e2026403118. [PMID: 34260376 PMCID: PMC8285897 DOI: 10.1073/pnas.2026403118] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
A significant unmet need for new contraceptive options for both women and men remains due to side-effect profiles, medical concerns, and the inconvenience of many currently available contraceptive products. Unfortunately, the development of novel nonsteroidal female contraceptive medicine has been stalled in the last couple of decades due to the lack of effective screening platforms. Drosophila utilizes conserved signaling pathways for follicle rupture, a final step in ovulation that is essential for female reproduction. Therefore, we explored the potential to use Drosophila as a model to screen compounds that could inhibit follicle rupture and be nonsteroidal contraceptive candidates. Using our ex vivo follicle rupture assay, we screened 1,172 Food and Drug Administration (FDA)-approved drugs and identified six drugs that could inhibit Drosophila follicle rupture in a dose-dependent manner. In addition, we characterized the molecular actions of these drugs in the inhibition of adrenergic signaling and follicle rupture. Furthermore, we validated that three of the four drugs consistently inhibited mouse follicle rupture in vitro and that two of them did not affect progesterone production. Finally, we showed that chlorpromazine, one of the candidate drugs, can significantly inhibit mouse follicle rupture in vivo. Our work suggests that Drosophila ovulation is a valuable platform for identifying lead compounds for nonsteroidal contraceptive development and highlights the potential of these FDA-approved drugs as novel nonsteroidal contraceptive agents.
Collapse
Affiliation(s)
- Kewa Jiang
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT 06269
| | - Jiyang Zhang
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL 60611
| | - Yuping Huang
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT 06269
| | - Yingzheng Wang
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ 08854
| | - Shuo Xiao
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ 08854
| | - M Kyle Hadden
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT 06269
| | - Teresa K Woodruff
- Department of Obstetrics and Gynecology, Northwestern University, Chicago, IL 60611
| | - Jianjun Sun
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT 06269;
- Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269
| |
Collapse
|
44
|
Wu T, Gao YY, Su J, Tang XN, Chen Q, Ma LW, Zhang JJ, Wu JM, Wang SX. Three-dimensional bioprinting of artificial ovaries by an extrusion-based method using gelatin-methacryloyl bioink. Climacteric 2021; 25:170-178. [PMID: 33993814 DOI: 10.1080/13697137.2021.1921726] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE The aim of this study was to design and fabricate a three-dimensional (3D) printed artificial ovary. METHODS We first compared the printability of gelatin-methacryloyl (GelMA), alginate and GelMA-alginate bioinks, of which GelMA was selected for further investigation. The swelling properties, degradation kinetics and shape fidelity of GelMA scaffolds were characterized by equilibrium swelling/lyophilization, collagenase processing and micro-computed tomography evaluation. Commercial ovarian tumor cell lines (COV434, KGN, ID8) and primary culture ovarian somatic cells were utilized to perform cell-laden 3D printing, and the results were evaluated by live/dead assays and TUNEL detection. Murine ovarian follicles were seeded in the ovarian scaffold and their diameters were recorded every day. Finally, in vitro maturation was performed, and the ovulated oocytes were collected and observed. RESULTS Our results indicated that GelMA was suitable for 3D printing fabrication. Its scaffolds performed well in terms of hygroscopicity, degradation kinetics and shape fidelity. The viability of ovarian somatic cells was lower than that of commercial cell lines, suggesting that extrusion-based 3D culture fabrication is not suitable for primary ovarian cells. Nevertheless, the GelMA-based 3D printing system provided an appropriate microenvironment for ovarian follicles, which successfully grew and ovulated in the scaffolds. Metaphase II oocytes were also observed after in vitro maturation. CONCLUSIONS The GelMA-based 3D printing culture system is a viable alternative option for follicular growth, development and transfer. Accordingly, it shows promise for clinical application in the treatment of female endocrine and reproductive conditions.
Collapse
Affiliation(s)
- T Wu
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Y Y Gao
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - J Su
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, China
| | - X N Tang
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Q Chen
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - L W Ma
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - J J Zhang
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - J M Wu
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, China
| | - S X Wang
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
45
|
Ferré-Pujol P, Otsuki J, Funahashi H, Nakatsuka M. The Thickness and Density of the Ovarian Tunica Albuginea Increases with Age in Transgender Patients. Reprod Sci 2021; 28:1339-1346. [PMID: 33398852 DOI: 10.1007/s43032-020-00390-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 11/08/2020] [Indexed: 10/22/2022]
Abstract
It is known that the extracellular matrix structure and composition changes with aging in many organs. Despite this, knowledge on how does the extracellular part of the ovary change with increasing age in women and how those changes might be related to women's loss of fertility is still lacking. For this, we propose that recurrent injury and repair events on the outermost layers of the ovary due to ovulation are partly responsible for those changes women experience with aging. The histological analysis of the ovaries from 18 female-to-male transgender patients revealed that the ovarian tunica albuginea (TA) increases its thickness and density correlatively with increasing age of the patient (r = 0.52 and r = 0.55, P < 0.05 respectively). The increase in thickness is independent of the total androgen dose received and occurs because of the appearance of defined fibrotic areas underneath the TA layer which increase the total distance of dense connective tissue from the ovarian surface. In conclusion, the ovarian TA increases in its thickness and density with aging because of the appearance of fibrotic areas underneath the layer in transgender patients. This fact might contribute to reduce oocyte quality and cause ovulation difficulties in older women.
Collapse
Affiliation(s)
- Pilar Ferré-Pujol
- Assisted Reproductive Technology (ART) Center, Okayama University, Okayama, Japan
| | - Junko Otsuki
- Assisted Reproductive Technology (ART) Center, Okayama University, Okayama, Japan.
- Graduate School of Environmental and Life Science, Okayama University, Okayama, Japan.
| | - Hiroaki Funahashi
- Assisted Reproductive Technology (ART) Center, Okayama University, Okayama, Japan.
- Graduate School of Environmental and Life Science, Okayama University, Okayama, Japan.
| | - Mikiya Nakatsuka
- Assisted Reproductive Technology (ART) Center, Okayama University, Okayama, Japan
- Okayama University Medical School, Okayama, Japan
| |
Collapse
|
46
|
Wu Y, Sun Z, Wang Y, Chen H, Bian J. Human dermal fibroblasts support the development of human primordial/primary follicles in a 3-dimensional alginate matrix culture system. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:868. [PMID: 34164502 PMCID: PMC8184424 DOI: 10.21037/atm-21-2125] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Alginate matrix 3-dimensional culture offers the opportunity for the development and maturation of human secondary follicles in vitro. However, alginate may not be the most suitable culture system for human primordial/primary follicles in vitro. Thus, the innovation of alginate matrix 3-dimensional culture systems for human primordial/primary follicles could hold promise as an ideal approach to restoring fertility. Methods We extracted primordial/primary follicles from ovarian tissues collected from patients with non-ovarian benign gynecological conditions. Fibroblasts were isolated from dermal tissue from 1 male patient who had undergone posthectomy. The isolated human follicles were randomly divided into 2 groups and encapsulated within fibroblast-alginate-hydrogels or alginate hydrogels. The survival and growth of human primordial/primary follicles were measured after 21 days of in vitro culture. Results The dermal fibroblasts in alginate hydrogel microcapsules were round in shape, and were distributed as uniform clouds on the surface and gaps of the alginate. After 21 days of culture, the survival rate of follicles in the fibroblast-alginate group was higher than that of the alginate group (P<0.05). The diameter of follicles in the fibroblast-alginate group and the alginate group after 21 days of culture was 152.80±13.64 and 129.14±9.95 μm, respectively (P<0.05). After 21-day culture, the mean cpm (log-converted) for 3H-thymidine incorporated by granulosa cells in the fibroblast-alginate and alginate groups was 6.87±0.24 and 4.63±0.38, respectively (P<0.05). After 21 days of culture, the messenger RNA expression levels of growth differentiation factor 9 (GDF9) and bone morphogenetic protein 15 (BMP15) were significantly higher in oocytes in fibroblast-alginate hydrogels than in those in alginate hydrogels (P<0.05). Conclusions Human fibroblasts are beneficial to the development of human follicles in 3-dimensional culture alginate gel systems over a long period of time. More studies are required to investigate the molecular biological mechanisms of human fibroblasts that promote follicle growth in vitro.
Collapse
Affiliation(s)
- Yuanyuan Wu
- Reproductive Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zili Sun
- Reproductive Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yu Wang
- Reproductive Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hong Chen
- Reproductive Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jiang Bian
- Department of Obstetrics and Gynecology, Shanghai Everjoy Medical Polyclinic, Shanghai, China
| |
Collapse
|
47
|
Telfer EE, Andersen CY. In vitro growth and maturation of primordial follicles and immature oocytes. Fertil Steril 2021; 115:1116-1125. [PMID: 33823993 DOI: 10.1016/j.fertnstert.2021.03.004] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/02/2021] [Accepted: 03/02/2021] [Indexed: 12/18/2022]
Abstract
Cryopreservation of ovarian tissue to preserve the fertility of girls and young women at high risk of sterility is now widely practiced. Pieces of cryopreserved ovarian cortex can be thawed and autografted to restore fertility, but because of the risks of reintroduction of the cancer, transplantation may not be possible for girls and women with blood-borne leukemias or cancers with a high risk of ovarian metastasis. Cryopreserved ovarian tissue contains mainly primordial follicles but also provides access to immature oocytes from small antral follicles, which may be matured in vitro to provide an additional source of mature oocytes. So in cases in which transplantation is contraindicated, fertility restoration could be safely achieved in the laboratory either by in vitro maturation (IVM) of oocytes aspirated from growing follicles or by the complete in vitro growth (IVG) and maturation (IVM) of primordial follicles to produce fertile metaphase II (MII) oocytes. The development of IVM and IVG methods to support all stages of oocytes available within ovarian tissue will maximize the potential for all patients undergoing fertility preservation.
Collapse
Affiliation(s)
- Evelyn E Telfer
- Institute of Cell Biology, The University of Edinburgh, Edinburgh EH8 8XE, Scotland.
| | - Claus Yding Andersen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, Faculty of Health and Medical Science, University of Copenhagen, DK-2100 Copenhagen, Denmark
| |
Collapse
|
48
|
Ouni E, Bouzin C, Dolmans MM, Marbaix E, Pyr Dit Ruys S, Vertommen D, Amorim CA. Spatiotemporal changes in mechanical matrisome components of the human ovary from prepuberty to menopause. Hum Reprod 2021; 35:1391-1410. [PMID: 32539154 DOI: 10.1093/humrep/deaa100] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 04/15/2020] [Accepted: 04/22/2020] [Indexed: 01/12/2023] Open
Abstract
STUDY QUESTION How do elastic matrisome components change during the lifetime of the human ovary? SUMMARY ANSWER The deposition and remodeling of mechanical matrisome components (collagen, elastin, elastin microfibril interface-located protein 1 (EMILIN-1), fibrillin-1 and glycosaminoglycans (GAGs)) that play key roles in signaling pathways related to follicle activation and development evolve in an age- and follicle stage-related manner. WHAT IS KNOWN ALREADY The mechanobiology of the human ovary and dynamic reciprocity that exists between ovarian cells and their microenvironment is of high importance. Indeed, while the localization of primordial follicles in the collagen-rich ovarian cortex offers a rigid physical environment that supports follicle architecture and probably plays a role in their survival, ovarian extracellular matrix (ECM) stiffness limits follicle expansion and hence oocyte maturation, maintaining follicles in their quiescent state. As growing follicles migrate to the medulla of the ovary, they encounter a softer, more pliant ECM, allowing expansion and development. Thus, changes in the rigidity of the ovarian ECM have a direct effect on follicle behavior. Evidence supporting a role for the physical environment in follicle activation was provided in clinical practice by ovarian tissue fragmentation, which promoted actin polymerization and disrupted ovarian Hippo signaling, leading to increased expression of downstream growth factors, promotion of follicle growth and generation of mature oocytes. STUDY DESIGN, SIZE, DURATION We investigated quantitative spatiotemporal changes in collagen, elastin, EMILIN-1, fibrillin-1 and GAGs from prepuberty to menopause, before conducting a closer analysis of the ECM surrounding follicles, from primordial to secondary stages, in both prepubertal and tissue from women of reproductive age. The study included ovarian tissue (cortex) from 68 patients of different ages: prepubertal (n = 16; mean age [±SD]=8 ± 2 years); reproductive (n = 21; mean age [±SD]=27 ± 4 years); menopausal with estrogen-based HRT (n = 7; mean age [±SD]=58 ± 4 years); and menopausal without HRT (n = 24; mean age [±SD]=61 ± 5 years). PARTICIPANTS/MATERIALS, SETTING, METHODS Quantitative investigations of collagen and GAG deposition in ovarian tissue throughout a woman's lifetime were conducted by analyzing brightfield images. Characteristic features of collagen fiber content were based on polarized light microscopy, since polarized light changes with fiber thickness. To evaluate the deposition and distribution of elastin, fibrillin-1 and EMILIN-1, multiplex immunofluorescence was used on at least three sections from each patient. Image processing and tailored bioinformatic analysis were applied to enable spatiotemporal quantitative evaluation of elastic system component deposition in the human ovary over its lifetime. MAIN RESULTS AND THE ROLE OF CHANCE While collagen levels increased with age, fibrillin-1 and EMILIN-1 declined. Interestingly, collagen and elastin reached their peak in reproductive-age women compared to prepubertal (P < 0.01; P = 0.262) and menopausal subjects with (P = 0.706; P < 0.01) and without (P = 0.987; P = 0.610) HRT, indicating a positive impact of secreted estrogen and hormone treatment on collagen and elastin preservation. Interestingly, HRT appears to affect elastin presence in ovarian tissue, since a significantly higher (P < 0.05) proportion of elastin was detected in biopsies from menopausal women taking HRT compared to those not. Higher GAG levels were found in adult ovaries compared to prepubertal ovaries (P < 0.05), suggesting changes in tissue ultrastructure and elasticity with age. In this context, elevated GAG values are suspected to participate in hampering formation of the fibrillin-1 network (r = -0.2475; P = 0.04687), which explains its decline over time. This decline partially accounts for the decrease in EMILIN-1 (r = 0.4149; P = 0.00059). Closer examination of the ECM surrounding follicles from the primordial to the secondary stage, both before and after puberty, points to high levels of mechanical stress placed on prepubertal follicles compared to the more compliant ECM around reproductive-age follicles, as suggested by the higher collagen levels and lower elastin content detected mainly around primordial (P < 0.0001; P < 0.0001, respectively) and primary (P < 0.0001; P < 0.001, respectively) follicles. Such a stiff niche is nonpermissive to prepubertal follicle activation and growth, and is more inclined to quiescence. LARGE SCALE DATA Not applicable. LIMITATIONS, REASONS FOR CAUTION The duration and form of administered HRT were not considered when studying the menopausal patient group undergoing treatment. Moreover, we cannot exclude interference from other nongynecological medications taken by the study patients on ovarian ECM properties since there is no information in the literature describing the impact of each medication on the ECM. Finally, since the ECM is by definition a very heterogeneous meshwork of proteins, the use of two-dimensional histology could be a limitation. Single time points on fixed tissues could also present limitations, since following ovary dynamics from prepuberty to menopause in the same patient is not feasible. WIDER IMPLICATIONS OF THE FINDINGS From a biomechanical perspective, our study revealed important changes to ECM properties dictating the mechanical features of ovarian tissue, in line with the existing literature. Our findings pave the way for possible therapeutic targets at the ECM level in the context of female fertility and ovarian rejuvenation, such as mechanical stimulation, antifibrotic treatments, and prevention or reversion of elastic ECM degradation. Our study also sheds light on the follicle-specific ECM composition that is dependent on follicle stage and age. These data will prove very useful in designing biomimetic scaffolds and tissue-engineered models like the artificial ovary. Indeed, they emphasize the importance of encapsulating each type of isolated follicle in an appropriate biomaterial that must replicate the corresponding functional perifollicular ECM and respect ovarian tissue heterogeneity in order to guarantee its biomimicry. STUDY FUNDING/COMPETING INTEREST(S) This study was supported by grants from the Fonds National de la Recherche Scientifique de Belgique (FNRS) (C.A.A. is an FRS-FNRS research associate; grant 5/4/150/5 awarded to M.M.D.) and the Université Catholique de Louvain (PhD grant 'Coopération au développement' awarded to E.O.). None of the authors have any competing interests to declare.
Collapse
Affiliation(s)
- E Ouni
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - C Bouzin
- IREC Imaging Platform (2IP), Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - M M Dolmans
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium.,Gynecology and Andrology Department, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - E Marbaix
- Pathology Department, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium.,Cell Biology Unit, de Duve Institute, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - S Pyr Dit Ruys
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - D Vertommen
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - C A Amorim
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium
| |
Collapse
|
49
|
Hakim BA, Tyagi V, Agnihotri SK, Nath A, Agrawal AK, Jain A, Singh D, Konwar R, Sachdev M. Electroporation of Mouse Follicles, Oocytes and Embryos without Manipulating Zona Pellucida. J Dev Biol 2021; 9:jdb9020013. [PMID: 33915920 PMCID: PMC8167572 DOI: 10.3390/jdb9020013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/23/2021] [Accepted: 03/26/2021] [Indexed: 11/16/2022] Open
Abstract
Electroporation is an effective technique of transfection, but its efficiency depends on the optimization of various parameters. In this study, a simplified and efficient method of gene manipulation was standardized through electroporation to introduce a recombinant green fluorescent protein (GFP) construct as well as RNA-inhibitors in intact mouse follicles, oocytes and early embryos, where various electroporation parameters like voltage, pulse number and pulse duration were standardized. Electroporated preantral follicles were cultured further in vitro to obtain mature oocytes and their viability was confirmed through the localization of a known oocyte maturation marker, ovastacin, which appeared to be similar to the in vivo-derived mature oocytes and thus proved the viability of the in vitro matured oocytes after electroporation. Standardized electroporation parameters, i.e., three pulses of 30 V for 1 millisecond at an interval of 10 s, were applied to manipulate the expression of mmu-miR-26a in preantral follicles through the electroporation of miR inhibitors and mimics. The TUNEL apoptosis assay confirmed the normal development of the electroporated embryos when compared to the normal embryos. Conclusively, for the first time, this study demonstrated the delivery of exogenous oligonucleotides into intact mouse follicles, oocytes and embryos without hampering their zona pellucida (ZP) and further development.
Collapse
Affiliation(s)
- Bilal Ahmad Hakim
- Division of Endocrinology, CSIR-Central Drug Research Institute (CDRI), Sector 10, Jankipuram Extension, Lucknow 226031, India; (B.A.H.); (V.T.); (S.K.A.); (A.N.); (A.K.A.); (A.J.); (D.S.); (R.K.)
| | - Vaishali Tyagi
- Division of Endocrinology, CSIR-Central Drug Research Institute (CDRI), Sector 10, Jankipuram Extension, Lucknow 226031, India; (B.A.H.); (V.T.); (S.K.A.); (A.N.); (A.K.A.); (A.J.); (D.S.); (R.K.)
| | - Saurabh Kumar Agnihotri
- Division of Endocrinology, CSIR-Central Drug Research Institute (CDRI), Sector 10, Jankipuram Extension, Lucknow 226031, India; (B.A.H.); (V.T.); (S.K.A.); (A.N.); (A.K.A.); (A.J.); (D.S.); (R.K.)
| | - Amar Nath
- Division of Endocrinology, CSIR-Central Drug Research Institute (CDRI), Sector 10, Jankipuram Extension, Lucknow 226031, India; (B.A.H.); (V.T.); (S.K.A.); (A.N.); (A.K.A.); (A.J.); (D.S.); (R.K.)
| | - Ankit Kumar Agrawal
- Division of Endocrinology, CSIR-Central Drug Research Institute (CDRI), Sector 10, Jankipuram Extension, Lucknow 226031, India; (B.A.H.); (V.T.); (S.K.A.); (A.N.); (A.K.A.); (A.J.); (D.S.); (R.K.)
| | - Ankita Jain
- Division of Endocrinology, CSIR-Central Drug Research Institute (CDRI), Sector 10, Jankipuram Extension, Lucknow 226031, India; (B.A.H.); (V.T.); (S.K.A.); (A.N.); (A.K.A.); (A.J.); (D.S.); (R.K.)
| | - Deependra Singh
- Division of Endocrinology, CSIR-Central Drug Research Institute (CDRI), Sector 10, Jankipuram Extension, Lucknow 226031, India; (B.A.H.); (V.T.); (S.K.A.); (A.N.); (A.K.A.); (A.J.); (D.S.); (R.K.)
| | - Rituraj Konwar
- Division of Endocrinology, CSIR-Central Drug Research Institute (CDRI), Sector 10, Jankipuram Extension, Lucknow 226031, India; (B.A.H.); (V.T.); (S.K.A.); (A.N.); (A.K.A.); (A.J.); (D.S.); (R.K.)
- Academy of Scientific and Innovative Research (AcSIR), New Delhi 110001, India
| | - Monika Sachdev
- Division of Endocrinology, CSIR-Central Drug Research Institute (CDRI), Sector 10, Jankipuram Extension, Lucknow 226031, India; (B.A.H.); (V.T.); (S.K.A.); (A.N.); (A.K.A.); (A.J.); (D.S.); (R.K.)
- Academy of Scientific and Innovative Research (AcSIR), New Delhi 110001, India
- Correspondence: ; Tel.: +91-9026561774
| |
Collapse
|
50
|
Creating an Artificial 3-Dimensional Ovarian Follicle Culture System Using a Microfluidic System. MICROMACHINES 2021; 12:mi12030261. [PMID: 33806282 PMCID: PMC7999445 DOI: 10.3390/mi12030261] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 02/25/2021] [Accepted: 02/28/2021] [Indexed: 12/12/2022]
Abstract
We hypothesized that the creation of a 3-dimensional ovarian follicle, with embedded granulosa and theca cells, would better mimic the environment necessary to support early oocytes, both structurally and hormonally. Using a microfluidic system with controlled flow rates, 3-dimensional two-layer (core and shell) capsules were created. The core consists of murine granulosa cells in 0.8 mg/mL collagen + 0.05% alginate, while the shell is composed of murine theca cells suspended in 2% alginate. Somatic cell viability tests and hormonal assessments (estradiol, progesterone, and androstenedione) were performed on days 1, 6, 13, 20, and 27. Confocal microscopy confirmed appropriate compartmentalization of fluorescently-labeled murine granulosa cells to the inner capsule and theca cells to the outer shell. Greater than 78% of cells present in capsules were alive up to 27 days after collection. Artificially constructed ovarian follicles exhibited intact endocrine function as evidenced by the production of estradiol, progesterone, and androstenedione. Oocytes from primary and early secondary follicles were successfully encapsulated, which maintained size and cellular compartmentalization. This novel microfluidic system successfully encapsulated oocytes from primary and secondary follicles, recapitulating the two-compartment system necessary for the development of the mammalian oocyte. Importantly, this microfluidic system can be easily adapted for sterile, high throughput applications.
Collapse
|