1
|
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
|
2
|
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
|
3
|
Nair R, Kasturi M, Mathur V, Seetharam RN, S Vasanthan K. Strategies for developing 3D printed ovarian model for restoring fertility. Clin Transl Sci 2024; 17:e13863. [PMID: 38955776 PMCID: PMC11219245 DOI: 10.1111/cts.13863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 05/16/2024] [Accepted: 05/31/2024] [Indexed: 07/04/2024] Open
Abstract
Ovaries play a crucial role in the regulation of numerous essential processes that occur within the intricate framework of female physiology. They are entrusted with the responsibility of both generating a new life and orchestrating a delicate hormonal symphony. Understanding their functioning is crucial for gaining insight into the complexities of reproduction, health, and fertility. In addition, ovaries secrete hormones that are crucial for both secondary sexual characteristics and the maintenance of overall health. A three-dimensional (3D) prosthetic ovary has the potential to restore ovarian function and preserve fertility in younger females who have undergone ovariectomies or are afflicted with ovarian malfunction. Clinical studies have not yet commenced, and the production of 3D ovarian tissue for human implantation is still in the research phase. The main challenges faced while creating a 3D ovary for in vivo implantation include sustenance of ovarian follicles, achieving vascular infiltration into the host tissue, and restoring hormone circulation. The complex ovarian microenvironment that is compartmentalized and rigid makes the biomimicking of the 3D ovary challenging in terms of biomaterial selection and bioink composition. The successful restoration of these properties in animal models has led to expectations for the development of human ovaries for implantation. This review article summarizes and evaluates the optimal 3D models of ovarian structures and their safety and efficacy concerns to provide concrete suggestions for future research.
Collapse
Affiliation(s)
- Ramya Nair
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher EducationManipalKarnatakaIndia
| | - Meghana Kasturi
- Department of Mechanical EngineeringUniversity of MichiganDearbornMichiganUSA
| | - Vidhi Mathur
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher EducationManipalKarnatakaIndia
| | - Raviraja N. Seetharam
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher EducationManipalKarnatakaIndia
| | - Kirthanashri S Vasanthan
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher EducationManipalKarnatakaIndia
| |
Collapse
|
4
|
Amjad F, Keshvari H, Dalman A, Montazeri L. In Vitro Development of Mouse Preantral Follicle with Using Amniotic Membrane Extract-Loaded Hydrogels. INTERNATIONAL JOURNAL OF FERTILITY & STERILITY 2024; 18:286-292. [PMID: 38973283 PMCID: PMC11245589 DOI: 10.22074/ijfs.2023.1990652.1443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Indexed: 07/09/2024]
Abstract
An artificial ovary based on the alginate (ALG) hydrogel has been widely implemented to preserve prepubertal female fertility. However, this platform is not fully capable of successful an ovary microenvironment simulation for follicle development, holding great potential for its improvement. Therefore, this experimental study aimed to evaluate the effect of an amniotic membrane extract (AME) -loaded hydrogel on the mouse preantral follicles in vitro development. In order to have better follicle development, first, the impact of different concentrations of follicle-stimulating hormone (FSH) was evaluated on the mouse preantral follicles encapsulated in ALG. Later, the appropriate dose was adjusted for the follicles encapsulated in the ALG-AME hydrogel. Results demonstrated that 100 mIU/ml FSH showed a significant follicle survival rate compared with 10 mIU/ml FSH (P=0.005). According to MTT assay finding, the rate of weight loss, and rheology evaluations, ALG containing 1 mg/ml AME was identified as an optimal sample of follicle culture instead of other AME concentrations. Follicle diameter significantly increased in the ALG-AME 1 hydrogel compared with the ALG control group without AME (P=0.027). The storage modulus of ALG-AME 1 was 773 Pa and retained the follicle morphology for 13 days. No statistically substantial difference was seen in survival, antrum cavity formation, and competent oocyte in terms of the normal chromosomal arrangement and meiotic spindle rate in comparison with the control group. It can be concluded that ALG-AME 1 could not significantly impact the mouse preantral follicle.
Collapse
Affiliation(s)
- Fatemeh Amjad
- Biomedical Engineering Department, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hamid Keshvari
- Biomedical Engineering Department, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran.
| | - Azam Dalman
- Department 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.
| |
Collapse
|
5
|
Hao J, Li T, Heinzelmann M, Moussaud-Lamodière E, Lebre F, Krjutškov K, Damdimopoulos A, Arnelo C, Pettersson K, Alfaro-Moreno E, Lindskog C, van Duursen M, Damdimopoulou P. Effects of chemical in vitro activation versus fragmentation on human ovarian tissue and follicle growth in culture. Hum Reprod Open 2024; 2024:hoae028. [PMID: 38803550 PMCID: PMC11128059 DOI: 10.1093/hropen/hoae028] [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: 11/28/2023] [Revised: 04/15/2024] [Indexed: 05/29/2024] Open
Abstract
STUDY QUESTION What is the effect of the chemical in vitro activation (cIVA) protocol compared with fragmentation only (Frag, also known as mechanical IVA) on gene expression, follicle activation and growth in human ovarian tissue in vitro? SUMMARY ANSWER Although histological assessment shows that cIVA significantly increases follicle survival and growth compared to Frag, both protocols stimulate extensive and nearly identical transcriptomic changes in cultured tissue compared to freshly collected ovarian tissue, including marked changes in energy metabolism and inflammatory responses. WHAT IS KNOWN ALREADY Treatments based on cIVA of the phosphatase and tensin homolog (PTEN)-phosphatidylinositol 3-kinase (PI3K) pathway in ovarian tissue followed by auto-transplantation have been administered to patients with refractory premature ovarian insufficiency (POI) and resulted in live births. However, comparable effects with mere tissue fragmentation have been shown, questioning the added value of chemical stimulation that could potentially activate oncogenic responses. STUDY DESIGN SIZE DURATION Fifty-nine ovarian cortical biopsies were obtained from consenting women undergoing elective caesarean section (C-section). The samples were fragmented for culture studies. Half of the fragments were exposed to bpV (HOpic)+740Y-P (Frag+cIVA group) during the first 24 h of culture, while the other half were cultured with medium only (Frag group). Subsequently, both groups were cultured with medium only for an additional 6 days. Tissue and media samples were collected for histological, transcriptomic, steroid hormone, and cytokine/chemokine analyses at various time points. PARTICIPANTS/MATERIALS SETTING METHODS Effects on follicles were evaluated by counting and scoring serial sections stained with hematoxylin and eosin before and after the 7-day culture. Follicle function was assessed by quantification of steroids by ultra-performance liquid chromatography tandem-mass spectrometry at different time points. Cytokines and chemokines were measured by multiplex assay. Transcriptomic effects were measured by RNA-sequencing (RNA-seq) of the tissue after the initial 24-h culture. Selected differentially expressed genes (DEGs) were validated by quantitative PCR and immunofluorescence in cultured ovarian tissue as well as in KGN cell (human ovarian granulosa-like tumor cell line) culture experiments. MAIN RESULTS AND THE ROLE OF CHANCE Compared to the Frag group, the Frag+cIVA group exhibited a significantly higher follicle survival rate, increased numbers of secondary follicles, and larger follicle sizes. Additionally, the tissue in the Frag+cIVA group produced less dehydroepiandrosterone compared to Frag. Cytokine measurement showed a strong inflammatory response at the start of the culture in both groups. The RNA-seq data revealed modest differences between the Frag+cIVA and Frag groups, with only 164 DEGs identified using a relaxed cut-off of false discovery rate (FDR) <0.1. Apart from the expected PI3K-protein kinase B (Akt) pathway, cIVA also regulated pathways related to hypoxia, cytokines, and inflammation. In comparison to freshly collected ovarian tissue, gene expression in general was markedly affected in both the Frag+cIVA and Frag groups, with a total of 3119 and 2900 DEGs identified (FDR < 0.001), respectively. The top enriched gene sets in both groups included several pathways known to modulate follicle growth such as mammalian target of rapamycin (mTOR)C1 signaling. Significant changes compared to fresh tissue were also observed in the expression of genes encoding for steroidogenesis enzymes and classical granulosa cell markers in both groups. Intriguingly, we discovered a profound upregulation of genes related to glycolysis and its upstream regulator in both Frag and Frag+cIVA groups, and these changes were further boosted by the cIVA treatment. Cell culture experiments confirmed glycolysis-related genes as direct targets of the cIVA drugs. In conclusion, cIVA enhances follicle growth, as expected, but the mechanisms may be more complex than PI3K-Akt-mTOR alone, and the impact on function and quality of the follicles after the culture period remains an open question. LARGE SCALE DATA Data were deposited in the GEO data base, accession number GSE234765. The code for sequencing analysis can be found in https://github.com/tialiv/IVA_project. LIMITATIONS REASONS FOR CAUTION Similar to the published IVA protocols, the first steps in our study were performed in an in vitro culture model where the ovarian tissue was isolated from the regulation of hypothalamic-pituitary-ovarian axis. Further in vivo experiments will be needed, for example in xeno-transplantation models, to explore the long-term impacts of the discovered effects. The tissue collected from patients undergoing C-section may not be comparable to tissue of patients with POI. WIDER IMPLICATIONS OF THE FINDINGS The general impact of fragmentation and short (24 h) in vitro culture on gene expression in ovarian tissue far exceeded the effects of cIVA. Yet, follicle growth was stimulated by cIVA, which may suggest effects on specific cell populations that may be diluted in bulk RNA-seq. Nevertheless, we confirmed the impact of cIVA on glycolysis using a cell culture model, suggesting impacts on cellular signaling beyond the PI3K pathway. The profound changes in inflammation and glycolysis following fragmentation and culture could contribute to follicle activation and loss in ovarian tissue culture, as well as in clinical applications, such as fertility preservation by ovarian tissue auto-transplantation. STUDY FUNDING/COMPETING INTERESTS This study was funded by research grants from European Union's Horizon 2020 Research and Innovation Programme (Project ERIN No. 952516, FREIA No. 825100), Swedish Research Council VR (2020-02132), StratRegen funding from Karolinska Institutet, KI-China Scholarship Council (CSC) Programme and the Natural Science Foundation of Hunan (2022JJ40782). International Iberian Nanotechnology Laboratory Research was funded by the European Union's H2020 Project Sinfonia (857253) and SbDToolBox (NORTE-01-0145-FEDER-000047), supported by Norte Portugal Regional Operational Programme (NORTE 2020), under the PORTUGAL 2020 Partnership Agreement, through the European Regional Development Fund. No competing interests are declared.
Collapse
Affiliation(s)
- Jie Hao
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, Changsha, P.R. China
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Tianyi Li
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Manuel Heinzelmann
- Department of Environment and Health, Amsterdam Institute for Life and Environment, Amsterdam, The Netherlands
| | - Elisabeth Moussaud-Lamodière
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Filipa Lebre
- Nanosafety Group, International Iberian Nanotechnology Laboratory, Braga, Portugal
| | - Kaarel Krjutškov
- Faculty of Medicine, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Competence Centre on Health Technologies, Tartu, Estonia
| | | | - Catarina Arnelo
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Karin Pettersson
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | | | - Cecilia Lindskog
- Department of Immunology, Genetics and Pathology, Cancer Precision Medicine Research Program, Uppsala University, Uppsala, Sweden
| | - Majorie van Duursen
- Department of Environment and Health, Amsterdam Institute for Life and Environment, Amsterdam, The Netherlands
| | - Pauliina Damdimopoulou
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
6
|
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
|
7
|
Richard S, Zhou Y, Jasoni CL, Pankhurst MW. Ovarian follicle size or growth rate can both be determinants of ovulatory follicle selection in mice†. Biol Reprod 2024; 110:130-139. [PMID: 37801701 PMCID: PMC10790341 DOI: 10.1093/biolre/ioad134] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 09/04/2023] [Accepted: 10/01/2023] [Indexed: 10/08/2023] Open
Abstract
The endocrinology regulating ovulation of the desired number of oocytes in the ovarian cycle is well described, particularly in mono-ovulatory species. Less is known about the characteristics that make one follicle suitable for ovulation while most other follicles die by atresia. Bromodeoxyuridine (BrdU) injection was used to characterize granulosa cell proliferation rates in developing ovarian follicles in the estrous cycle of mice. This methodology allowed identification of follicle diameters of secondary (80-130 μm), follicle-stimulating hormone (FSH)-sensitive (130-170 μm), FSH-dependent (170-350 μm), and preovulatory (>350 μm) follicles. Few preovulatory-sized follicles were present in the ovaries of mice at estrus, the beginning of the cycle. Progressive increases were seen at metestrus and diestrus, when full accumulation of the preovulatory cohort (~10 follicles) occurred. BrdU pulse-chase studies determined granulosa cell proliferation rates in the 24-48 h before the follicle reached the preovulatory stage. This showed that slow-growing follicles were not able to survive to the preovulatory stage. Mathematical modeling of follicle growth rates determined that the largest follicles at the beginning of the cycle had the greatest chance of becoming preovulatory. However, smaller follicles could enter the preovulatory follicle pool if low numbers of large antral follicles were present at the beginning of the cycle. In this instance, rapidly growing follicles had a clear selection advantage. The developing follicle pool displays heterogeneity in granulosa cell proliferation rates, even among follicles at the same stage of development. This parameter appears to influence whether a follicle can ovulate or become atretic.
Collapse
Affiliation(s)
- Sharon Richard
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Yiran Zhou
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Christine L Jasoni
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- Centre for Neuroendocrinology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Michael W Pankhurst
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
8
|
Shi Y, Guo Y, Zhou J, Cui G, Cheng J, Wu Y, Zhao Y, Fang L, Han X, Yang Y, Sun Y. A spatiotemporal gene expression and cell atlases of the developing rat ovary. Cell Prolif 2023; 56:e13516. [PMID: 37309718 PMCID: PMC10693188 DOI: 10.1111/cpr.13516] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 06/14/2023] Open
Abstract
Normal ovarian development is necessary for the production of healthy oocytes. However, the characteristics of oocytes development at different stages and the regulatory relationship between oocytes and somatic cells remain to be fully explained. Here, we combined scRNA-seq and spatial transcriptomic sequencing to profile the transcriptomic atlas of developing ovarian of the rat. We identified four components from developing granulosa cells including cumulus, primitive, mural, and luteal cells, and constructed their differential transcriptional regulatory networks. Several novel growth signals from oocytes to cumulus cells were identified, such as JAG1-NOTCH2 and FGF9-FGFR2. Moreover, we observed three cumulus sequential phases during follicle development determined by the key transcriptional factors in each cumulus phase (Bckaf1, Gata6, Cebpb, etc.), as well as the potential pinpointed roles of macrophages in luteal regression. Altogether, the single-cell spatial transcriptomic profile of the ovary provides not only a new research dimension for temporal and spatial analysis of ovary development, but also valuable data resources and a research basis for in-depth excavation of the mechanisms of mammalian ovary development.
Collapse
Affiliation(s)
- Yong Shi
- Henan Key Laboratory of Reproduction and Genetics, Center for Reproductive MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Academy of medical sciencesZhengzhou UniversityZhengzhouChina
| | - Yanjie Guo
- Henan Key Laboratory of Reproduction and Genetics, Center for Reproductive MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jiayi Zhou
- Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, College of Future Technology, Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
- China National Center for BioinformationBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Guanshen Cui
- Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, College of Future Technology, Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
- China National Center for BioinformationBeijingChina
| | - Jung‐Chien Cheng
- Henan Key Laboratory of Reproduction and Genetics, Center for Reproductive MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Ying Wu
- Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, College of Future Technology, Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
- China National Center for BioinformationBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yong‐Liang Zhao
- Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, College of Future Technology, Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
- China National Center for BioinformationBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Lanlan Fang
- Henan Key Laboratory of Reproduction and Genetics, Center for Reproductive MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Xiao Han
- Henan Key Laboratory of Reproduction and Genetics, Center for Reproductive MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, College of Future Technology, Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
- China National Center for BioinformationBeijingChina
| | - Yun‐Gui Yang
- Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, College of Future Technology, Beijing Institute of GenomicsChinese Academy of SciencesBeijingChina
- China National Center for BioinformationBeijingChina
- University of Chinese Academy of SciencesBeijingChina
- Institute of Stem Cell and RegenerationChinese Academy of SciencesBeijingChina
| | - Yingpu Sun
- Henan Key Laboratory of Reproduction and Genetics, Center for Reproductive MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
9
|
Converse A, Liu Z, Patel JC, Shakyawar S, Guda C, Bousfield GR, Kumar TR, Duncan FE. Oocyte quality is enhanced by hypoglycosylated FSH through increased cell-to-cell interaction during mouse follicle development. Development 2023; 150:dev202170. [PMID: 37870089 PMCID: PMC10651093 DOI: 10.1242/dev.202170] [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: 07/09/2023] [Accepted: 10/10/2023] [Indexed: 10/24/2023]
Abstract
Macroheterogeneity in follicle-stimulating hormone (FSH) β-subunit N-glycosylation results in distinct FSH glycoforms. Hypoglycosylated FSH21 is the abundant and more bioactive form in pituitaries of females under 35 years of age, whereas fully glycosylated FSH24 is less bioactive and increases with age. To investigate whether the shift in FSH glycoform abundance contributes to the age-dependent decline in oocyte quality, the direct effects of FSH glycoforms on folliculogenesis and oocyte quality were determined using an encapsulated in vitro mouse follicle growth system. Long-term culture (10-12 days) with FSH21 (10 ng/ml) enhanced follicle growth, estradiol secretion and oocyte quality compared with FSH24 (10 ng/ml) treatment. FSH21 enhanced establishment of transzonal projections, gap junctions and cell-to-cell communication within 24 h in culture. Transient inhibition of FSH21-mediated bidirectional communication abrogated the positive effects of FSH21 on follicle growth, estradiol secretion and oocyte quality. Our data indicate that FSH21 promotes folliculogenesis and oocyte quality in vitro by increasing cell-to-cell communication early in folliculogenesis, and that the shift in in vivo abundance from FSH21 to FSH24 with reproductive aging may contribute to the age-dependent decline in oocyte quality.
Collapse
Affiliation(s)
- Aubrey Converse
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Zhenghui Liu
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jai C. Patel
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Sushil Shakyawar
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Chittibabu Guda
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - George R. Bousfield
- Department of Biological Sciences, Wichita State University, Wichita, KS 67260, USA
| | - T. Rajendra Kumar
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Francesca E. Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
10
|
Torkzadeh T, Asadi Z, Jafari Atrabi M, Eivazkhani F, Khodadi M, Hajiaghalou S, Akbarinejad V, Fathi R. Optimisation of hormonal treatment to improve follicular development in one-day-old mice ovaries cultured under in vitro condition. Reprod Fertil Dev 2023; 35:733-749. [PMID: 37995332 DOI: 10.1071/rd23027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 10/30/2023] [Indexed: 11/25/2023] Open
Abstract
CONTEXT Base medium containing knock-out serum replacement (KSR) has been found to support formation and maintenance of follicles in one-day-old mice ovaries, but has not been shown to properly support activation and growth of primordial follicles. AIMS The present study was conducted to tailor the hormonal content of base medium containing KSR to enhance development of primordial follicles in neonatal ovaries. METHODS One-day-old mice ovaries were initially cultured with base medium for four days, and then, different hormonal treatments were added to the culture media and the culture was proceeded for four additional days until day eight. Ovaries were collected for histological and molecular assessments on days four and eight. KEY RESULTS In experiment I, the main and interactive effects of FSH and testosterone were investigated and FSH promoted activation of primordial follicles and development of primary and preantral follicles, and upregulated genes of phosphoinositide 3-kinase (Pi3k ), KIT ligand (Kitl ), growth differentiation factor 9 (Gdf9 ) and follicle stimulating hormone receptor (Fshr ) (P Bmp15 ), Connexin-43 (Cx43 ) and luteinising hormone and choriogonadotropin receptor (Lhcgr ) (P P Lhcgr (P P >0.05). CONCLUSIONS Supplementation of culture medium containing KSR with gonadotropins, particularly hMG, could improve follicular growth and expression of factors regulating follicular development. IMPLICATIONS This study was a step forward in formulating an optimal medium for development of follicles in cultured one-day-old mice ovaries.
Collapse
Affiliation(s)
- Tahoura Torkzadeh
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Zahra Asadi
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran; and Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73014, USA
| | - Mohammad Jafari Atrabi
- Institute of Pharmacology and Toxicology, University Medical Center, Georg August University, Göttingen, Germany; and Platform Degenerative Diseases, German Primate Center, Leibniz Institute for Primate Research (DPZ), Göttingen, Germany
| | - Farideh Eivazkhani
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Maryam Khodadi
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Samira Hajiaghalou
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Vahid Akbarinejad
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Rouhollah Fathi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| |
Collapse
|
11
|
Zhu Q, Li Y, Ma J, Ma H, Liang X. Potential factors result in diminished ovarian reserve: a comprehensive review. J Ovarian Res 2023; 16:208. [PMID: 37880734 PMCID: PMC10598941 DOI: 10.1186/s13048-023-01296-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 10/07/2023] [Indexed: 10/27/2023] Open
Abstract
The ovarian reserve is defined as the quantity of oocytes stored in the ovary or the number of oocytes that can be recruited. Ovarian reserve can be affected by many factors, including hormones, metabolites, initial ovarian reserve, environmental problems, diseases, and medications, among others. With the trend of postponing of pregnancy in modern society, diminished ovarian reserve (DOR) has become one of the most common challenges in current clinical reproductive medicine. Attributed to its unclear mechanism and complex clinical features, it is difficult for physicians to administer targeted treatment. This review focuses on the factors associated with ovarian reserve and discusses the potential influences and pathogenic factors that may explain the possible mechanisms of DOR, which can be improved or built upon by subsequent researchers to verify, replicate, and establish further study findings, as well as for scientists to find new treatments.
Collapse
Affiliation(s)
- Qinying Zhu
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Yi Li
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Jianhong Ma
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Hao Ma
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Xiaolei Liang
- Department of Obstetrics and Gynecology, Key Laboratory for Gynecologic Oncology Gansu Province, The First Hospital of Lanzhou University, No.1, Donggangxi Rd, Chengguan District, 730000, Lanzhou, China.
| |
Collapse
|
12
|
Fraire-Zamora JJ, Sharma K, Ammar OF, Massarotti C, Ali ZE, Telfer EE, Williams S, Ata B, Liperis G. Mind the gap: deciphering the role of anti-Müllerian hormone in follicular development-from animal studies toward clinical application. Hum Reprod 2023; 38:1231-1234. [PMID: 37075310 DOI: 10.1093/humrep/dead075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Indexed: 04/21/2023] Open
Affiliation(s)
| | - Kashish Sharma
- HealthPlus Fertility and Women's Health Centre, Abu Dhabi, UAE
| | - Omar F Ammar
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, UK
| | - Claudia Massarotti
- Academic Unit of Obstetrics and Gynecology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- DINOGMI Department, University of Genova, Genova, Italy
| | - Zoya E Ali
- Research & Development Department, Hertility Health Limited, London, UK
| | - Evelyn E Telfer
- University of Edinburgh Institute of Cell Biology, Edinburgh, UK
| | - Suzannah Williams
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, UK
| | - Baris Ata
- Obstetrics and Gynecology Department, Koc University, Istanbul, Turkey
- ART Fertility Clinics, Dubai, United Arab Emirates
| | | |
Collapse
|
13
|
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
|
14
|
Han J, Zhang N, Cao Q, Shi X, Wang C, Rui X, Ding J, Zhao C, Zhang J, Ling X, Li H, Guan Y, Meng Q, Huo R. NLRP7 participates in the human subcortical maternal complex and its variants cause female infertility characterized by early embryo arrest. J Mol Med (Berl) 2023:10.1007/s00109-023-02322-7. [PMID: 37148315 DOI: 10.1007/s00109-023-02322-7] [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: 10/12/2022] [Revised: 04/10/2023] [Accepted: 04/12/2023] [Indexed: 05/08/2023]
Abstract
Successful human reproduction requires normal oocyte maturation, fertilization, and early embryo development. Early embryo arrest is a common phenomenon leading to female infertility, but the genetic basis is largely unknown. NLR family pyrin domain-containing 7 (NLRP7) is a member of the NLRP subfamily. Previous studies have shown that variants of NLRP7 are one of the crucial causes of female recurrent hydatidiform mole, but whether NLRP7 variants can directly affect early embryo development is unclear. We performed whole-exome sequencing in patients who experienced early embryo arrest, and five heterozygous variants (c.251G > A, c.1258G > A, c.1441G > A, c. 2227G > A, c.2323C > T) of NLRP7 were identified in affected individuals. Plasmids of NLRP7 and subcortical maternal complex components were overexpressed in 293 T cells, and Co-IP experiments showed that NLRP7 interacted with NLRP5, TLE6, PADI6, NLRP2, KHDC3L, OOEP, and ZBED3. Injecting complementary RNAs in mouse oocytes and early embryos showed that NLRP7 variants influenced the oocyte quality and some of the variants significantly affected early embryo development. These findings contribute to our understanding of the role of NLRP7 in human early embryo development and provide a new genetic marker for clinical early embryo arrest patients. KEY MESSAGES: Five heterozygous variants of NLRP7 (c.1441G > A; 2227G > A; c.251G > A; c.1258G > A; c.2323C > T) were identified in five infertile patients who experienced early embryo arrest. NLRP7 is a component of human subcortical maternal complex. NLRP7 variants lead to poor quality of oocytes and early embryo development arrest. This study provides a new genetic marker for clinical early embryo arrest patients.
Collapse
Affiliation(s)
- Jian Han
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Nana Zhang
- Center for Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qiqi Cao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Xiaodan Shi
- Department of Reproductive Medicine, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Congjing Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Ximan Rui
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Jie Ding
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
- Reproductive Genetic Center, Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Chun Zhao
- Department of Reproductive Medicine, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Junqiang Zhang
- Department of Reproductive Medicine, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Xiufeng Ling
- Department of Reproductive Medicine, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Hong Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
- Reproductive Genetic Center, Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Yichun Guan
- Center for Reproductive Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Qingxia Meng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China.
- Reproductive Genetic Center, Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China.
| | - Ran Huo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China.
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
15
|
Costa CB, Fair T, Seneda MM. Review: Environment of the ovulatory follicle: modifications and use of biotechnologies to enhance oocyte competence and increase fertility in cattle. Animal 2023; 17 Suppl 1:100866. [PMID: 37567670 DOI: 10.1016/j.animal.2023.100866] [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: 11/23/2022] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 08/13/2023] Open
Abstract
The oocyte is the basis of life, supporting development from a fertilized cell to an independent multicellular organism. The oocyte's competence to drive the first cell cycles postfertilization are critical to embryonic survival and subsequent successful pregnancy. Coupled with the complex processes of follicle assembly, activation, differentiation, growth, and terminal maturation, oocyte developmental competence is gradually acquired during oocyte growth and meiotic maturation. Most reproduction management technologies and interventions are centered around these highly coordinated processes, targeting the ovarian follicle and the oocyte within. Thus, our objective was to highlight key aspects of oocyte and follicle development in cattle, and to discuss recent advances in oocyte and follicle-centered reproductive biotechnologies.
Collapse
Affiliation(s)
- Camila Bortoliero Costa
- Department of Biological Sciences, School of Sciences and Languages, São Paulo State University (UNESP), Campus Assis, São Paulo, Brazil; Graduate Program in Pharmacology and Biotechnology, Institute of Biosciences, UNESP, Botucatu, São Paulo, Brazil
| | - Trudee Fair
- School of Agriculture and Food Science, University College Dublin, Ireland
| | - Marcelo M Seneda
- State University of Londrina (UEL), Laboratory of Animal Reproduction, Londrina, PR, Brazil.
| |
Collapse
|
16
|
Jitjumnong J, Tang PC. Bone Morphogenetic Protein 15 (BMP-15) Improves In Vitro Mouse Folliculogenesis. Animals (Basel) 2023; 13:ani13060980. [PMID: 36978521 PMCID: PMC10044016 DOI: 10.3390/ani13060980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/16/2023] [Accepted: 03/01/2023] [Indexed: 03/10/2023] Open
Abstract
Multilayered secondary follicles were encapsulated in a 0.5% alginate matrix and cultured in a 3D culture system supplemented with bone morphogenetic protein 15 (BMP-15; 15 ng/mL) for 12 days. The in vitro development of ovarian follicles was evaluated. On day 12, the follicle diameter, follicle survival rate, and antrum formation rate were significantly higher for follicles cultured in BMP-15-supplemented medium than those cultured in regular medium. The percentage of ovulated metaphase II oocytes retrieved from follicles cultured in BMP-15-supplemented medium was greater than that of oocytes retrieved from follicles cultured in regular medium. The secretion of P4 was significantly higher on days 6, 8, and 10 in follicles cultured in BMP-15-supplemented medium. The result for E2 tended toward significance on day 12. Intracellular reactive oxygen species levels were higher and glutathione levels were lower in mature oocytes from the in vitro culture than in mature oocytes from an in vivo control. A 3D culture system using an alginate matrix and supplemented with BMP-15 effectively improves the outcomes of in vitro ovarian follicle culture.
Collapse
Affiliation(s)
- Jakree Jitjumnong
- Department of Animal Science, National Chung Hsing University, Taichung 40227, Taiwan
| | - Pin-Chi Tang
- Department of Animal Science, National Chung Hsing University, Taichung 40227, Taiwan
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung 40227, Taiwan
- Correspondence: ; Tel.: +886-4-2284-0365 (ext. 222); Fax: +886-4-2286-0265
| |
Collapse
|
17
|
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
|
18
|
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
|
19
|
Marchais M, Gilbert I, Bastien A, Macaulay A, Robert C. Mammalian cumulus-oocyte complex communication: a dialog through long and short distance messaging. J Assist Reprod Genet 2022; 39:1011-1025. [PMID: 35499777 PMCID: PMC9107539 DOI: 10.1007/s10815-022-02438-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 02/13/2022] [Indexed: 12/13/2022] Open
Abstract
Communications are crucial to ovarian follicle development and to ovulation, and while both folliculogenesis and oogenesis are distinct processes, they share highly interdependent signaling pathways. Signals from distant organs such as the brain must be processed and compartments within the follicle have to be synchronized. The hypothalamic–pituitary–gonadal (HPG) axis relies on long-distance signalling analogous to wireless communication by which data is disseminated in the environment and cells equipped with the appropriate receptors receive and interpret the messages. In contrast, direct cell-to-cell transfer of molecules is a very targeted, short distance messaging system. Numerous signalling pathways have been identified and proven to be essential for the production of a developmentally competent egg. The development of the cumulus-oocyte complex relies largely on short distance communications or direct transfer type via extensions of corona radiata cells through the zona pellucida. The type of information transmitted through these transzonal projections is still largely uncharacterized. This review provides an overview of current understanding of the mechanisms by which the gamete receives and transmits information within the follicle. Moreover, it highlights the fact that in addition to the well-known systemic long-distance based communications from the HPG axis, these mechanisms acting more locally should also be considered as important targets for controlling/optimizing oocyte quality.
Collapse
Affiliation(s)
- Mathilde Marchais
- Département des sciences animales, Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle (CRDSI), Réseau Québécois en Reproduction (RQR), Pavillon Paul Comtois, Université Laval, Québec, QC, Canada
| | - Isabelle Gilbert
- Département des sciences animales, Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle (CRDSI), Réseau Québécois en Reproduction (RQR), Pavillon Paul Comtois, Université Laval, Québec, QC, Canada
| | - Alexandre Bastien
- Département des sciences animales, Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle (CRDSI), Réseau Québécois en Reproduction (RQR), Pavillon Paul Comtois, Université Laval, Québec, QC, Canada
| | - Angus Macaulay
- Département des sciences animales, Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle (CRDSI), Réseau Québécois en Reproduction (RQR), Pavillon Paul Comtois, Université Laval, Québec, QC, Canada
| | - Claude Robert
- Département des sciences animales, Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle (CRDSI), Réseau Québécois en Reproduction (RQR), Pavillon Paul Comtois, Université Laval, Québec, QC, Canada.
| |
Collapse
|
20
|
Liu X, Mai H, Chen P, Zhang Z, Wu T, Chen J, Sun P, Zhou C, Liang X, Huang R. Comparative analyses in transcriptome of human granulosa cells and follicular fluid micro-environment between poor ovarian responders with conventional controlled ovarian or mild ovarian stimulations. Reprod Biol Endocrinol 2022; 20:54. [PMID: 35313911 PMCID: PMC8935846 DOI: 10.1186/s12958-022-00926-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/07/2022] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Both mild and conventional controlled ovarian stimulation are the frequently used protocols for poor ovarian responders. However, there are some debates about which treatment is better. Moreover, little is known about the follicular physiology after the two ovarian stimulation protocols. This study was intended to investigate the features in granulosa cells and follicular fluid micro-environment after the two different ovarian stimulation protocols in poor responders. METHODS Granulosa cells RNA were sequenced using Illumina Hiseq technology. Specific differently expressed genes and proteins were verified by real-time quantitative PCR and Western blot analysis. Moreover, hormone and cytokine concentrations in the follicular fluid were measured by electrochemiluminescence immunoassay and enzyme-linked immunoabsorbent assay. The correlation between the results of molecular experiments and the laboratory outcomes were analyzed by Spearman correlation analysis. RESULTS The differentially expressed genes between the two groups were involved in 4 signaling pathways related to the follicular development; three proteins pertinent to the TGF-β signaling pathway were expressed differently in granulosa cells between the two, and the constituents in the follicular fluid were also different. Further, a correlation between the TGF-β signaling pathway and the good-quality embryo was observed. CONCLUSIONS The present study made a comparison for the first time in the transcriptome of human granulosa cells and the follicular fluid micro-environment between poor responders with the conventional controlled ovarian stimulation or the mild ovarian stimulation, showing that the TGF-β signaling pathway may correlate with the good-quality of embryos in the mild group, which may be instrumental to the choice of optimal management for IVF patients.
Collapse
Affiliation(s)
- Xiaoping Liu
- Reproductive Medicine Research Center, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Huisi Mai
- Reproductive Medicine Research Center, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Panyu Chen
- Reproductive Medicine Research Center, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhiqiang Zhang
- Reproductive Medicine Research Center, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Taibao Wu
- Reproductive Medicine Research Center, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jianhui Chen
- Reproductive Medicine Research Center, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Peng Sun
- Reproductive Medicine Research Center, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Chuanchuan Zhou
- Reproductive Medicine Research Center, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiaoyan Liang
- Reproductive Medicine Research Center, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Rui Huang
- Reproductive Medicine Research Center, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
21
|
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
|
22
|
Robert C. Nurturing the egg: the essential connection between cumulus cells and the oocyte. Reprod Fertil Dev 2021; 34:149-159. [PMID: 35231386 DOI: 10.1071/rd21282] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The determinants of oocyte quality remain uncertain. Under suitable conditions, which have yet to be defined, the gamete grows and acquires the competence to resume meiosis, be fertilised and undergo embryonic development at least beyond genome activation, after which the blastomere is autonomous enough to adapt to the specificity of its environment. This review describes the central role played by the oocyte in reproductive success and how communication between cumulus cells and the oocyte are essential to proper oogenesis and the quality of the resulting gamete. While most attempts to improve oocyte quality have been directed at gonadotrophin-based systemic endocrine signalling, it is proposed that parallel control of fertility may act locally within ovarian follicles through intimate cooperation between somatic cells and the oocyte via the network of transzonal projections. This intercellular communication may prove to be more sensitive to environmental conditions than systemic endocrine signalling, which is essential for many non-reproductive tissues.
Collapse
Affiliation(s)
- Claude Robert
- Département des sciences animales, Faculté des sciences de l'agriculture et de l'alimentation, Université Laval, Québec, QC, Canada
| |
Collapse
|
23
|
Fadeeva IV, Trofimchuk ES, Forysenkova AA, Ahmed AI, Gnezdilov OI, Davydova GA, Kozlova SG, Antoniac A, Rau JV. Composite Polyvinylpyrrolidone-Sodium Alginate-Hydroxyapatite Hydrogel Films for Bone Repair and Wound Dressings Applications. Polymers (Basel) 2021; 13:polym13223989. [PMID: 34833286 PMCID: PMC8621946 DOI: 10.3390/polym13223989] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/09/2021] [Accepted: 11/15/2021] [Indexed: 12/21/2022] Open
Abstract
Today, the synthesis of biocompatible and bioresorbable composite materials such as “polymer matrix-mineral constituent,” which stimulate the natural growth of living tissues and the restoration of damaged parts of the body, is one of the challenging problems in regenerative medicine. In this study, composite films of bioresorbable polymers of polyvinylpyrrolidone (PVP) and sodium alginate (SA) with hydroxyapatite (HA) were obtained. HA was introduced by two different methods. In one of them, it was synthesized in situ in a solution of polymer mixture, and in another one, it was added ex situ. Phase composition, microstructure, swelling properties and biocompatibility of films were investigated. The crosslinked composite PVP-SA-HA films exhibit hydrogel swelling characteristics, increasing three times in mass after immersion in a saline solution. It was found that composite PVP-SA-HA hydrogel films containing HA synthesized in situ exhibited acute cytotoxicity, associated with the presence of HA synthesis reaction byproducts—ammonia and ammonium nitrate. On the other hand, the films with HA added ex situ promoted the viability of dental pulp stem cells compared to the films containing only a polymer PVP-SA blend. The developed composite hydrogel films are recommended for such applications, such as membranes in osteoplastic surgery and wound dressing.
Collapse
Affiliation(s)
- Inna V. Fadeeva
- Baikov Institute of Metallurgy and Material Science RAS, Leninsky, 49, 119334 Moscow, Russia;
- Correspondence: (I.V.F.); (J.V.R.)
| | - Elena S. Trofimchuk
- Department of High-Molecular Compounds, Lomonosov Moscow State University, GSP-1, 1-3 Leninskiye Gory, 119991 Moscow, Russia;
| | - Anna A. Forysenkova
- Baikov Institute of Metallurgy and Material Science RAS, Leninsky, 49, 119334 Moscow, Russia;
| | - Abdulrahman I. Ahmed
- Department of Physics, Kazan Federal University, Kremlevskaya 18, 420008 Kazan, Russia; (A.I.A.); (O.I.G.)
- Department of Physics, University of Al-Hamadaniya, Mosul 41001, Iraq
| | - Oleg I. Gnezdilov
- Department of Physics, Kazan Federal University, Kremlevskaya 18, 420008 Kazan, Russia; (A.I.A.); (O.I.G.)
| | - Galina A. Davydova
- Institute of Theoretical and Experimental Biophysics of RAS, Institutskaya 3, 142290 Pushchino, Moscow reg., Russia;
- National Medical Research Center of Obstetrics, Gynecology and Perinatology, Academician Oparin Str., 117997 Moscow, Russia
| | - Svetlana G. Kozlova
- Department of Natural Science, Novosibirsk State University, Pirogova Street 2, 630090 Novosibirsk, Russia;
| | - Aurora Antoniac
- Department of Metallic Materials Science and Physical Metallurgy, University Politehnica of Bucharest, Street Splaiul Independentei, 060042 Bucharest, Romania;
| | - Julietta V. Rau
- Istituto di Struttura della Materia, Consiglio Nazionale delle Ricerche (ISM-CNR), Via del Fosso del Cavaliere, 00133 Rome, Italy
- Department of Analytical, Physical and Colloid Chemistry, I.M. Sechenov First Moscow State Medical University, Trubetskaya Street, Build. 8/2, 119991 Moscow, Russia
- Correspondence: (I.V.F.); (J.V.R.)
| |
Collapse
|
24
|
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
|
25
|
Dadashzadeh A, Moghassemi S, Shavandi A, Amorim CA. A review on biomaterials for ovarian tissue engineering. Acta Biomater 2021; 135:48-63. [PMID: 34454083 DOI: 10.1016/j.actbio.2021.08.026] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/26/2021] [Accepted: 08/18/2021] [Indexed: 12/19/2022]
Abstract
Considerable challenges in engineering the female reproductive tissue are the follicle's unique architecture, the need to recapitulate the extracellular matrix, and tissue vascularization. Over the years, various strategies have been developed for preserving fertility in women diagnosed with cancer, such as embryo, oocyte, or ovarian tissue cryopreservation. While autotransplantation of cryopreserved ovarian tissue is a viable choice to restore fertility in prepubertal girls and women who need to begin chemo- or radiotherapy soon after the cancer diagnosis, it is not suitable for all patients due to the risk of having malignant cells present in the ovarian fragments in some types of cancer. Advances in tissue engineering such as 3D printing and ovary-on-a-chip technologies have the potential to be a translational strategy for precisely recapitulating normal tissue in terms of physical structure, vascularization, and molecular and cellular spatial distribution. This review first introduces the ovarian tissue structure, describes suitable properties of biomaterials for ovarian tissue engineering, and highlights recent advances in tissue engineering for developing an artificial ovary. STATEMENT OF SIGNIFICANCE: The increase of survival rates in young cancer patients has been accompanied by a rise in infertility/sterility in cancer survivors caused by the gonadotoxic effect of some chemotherapy regimens or radiotherapy. Such side-effect has a negative impact on these patients' quality of life as one of their main concerns is generating biologically related children. To aid female cancer patients, several research groups have been resorting to tissue engineering strategies to develop an artificial ovary. In this review, we discuss the numerous biomaterials cited in the literature that have been tested to encapsulate and in vitro culture or transplant isolated preantral follicles from human and different animal models. We also summarize the recent advances in tissue engineering that can potentially be optimal strategies for developing an artificial ovary.
Collapse
|
26
|
Bodke VV, Burdette JE. Advancements in Microfluidic Systems for the Study of Female Reproductive Biology. Endocrinology 2021; 162:6225875. [PMID: 33852726 PMCID: PMC8571709 DOI: 10.1210/endocr/bqab078] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Indexed: 12/11/2022]
Abstract
The female reproductive tract is a highly complex physiological system that consists of the ovaries, fallopian tubes, uterus, cervix, and vagina. An enhanced understanding of the molecular, cellular, and genetic mechanisms of the tract will allow for the development of more effective assisted reproductive technologies, therapeutics, and screening strategies for female specific disorders. Traditional 2-dimensional and 3-dimensional static culture systems may not always reflect the cellular and physical contexts or physicochemical microenvironment necessary to understand the dynamic exchange that is crucial for the functioning of the reproductive system. Microfluidic systems present a unique opportunity to study the female reproductive tract, as these systems recapitulate the multicellular architecture, contacts between different tissues, and microenvironmental cues that largely influence cell structure, function, behavior, and growth. This review discusses examples, challenges, and benefits of using microfluidic systems to model ovaries, fallopian tubes, endometrium, and placenta. Additionally, this review also briefly discusses the use of these systems in studying the effects of endocrine disrupting chemicals and diseases such as ovarian cancer, preeclampsia, and polycystic ovarian syndrome.
Collapse
Affiliation(s)
- Vedant V Bodke
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago 60607, USA
| | - Joanna E Burdette
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago 60607, USA
- Correspondence: Joanna E. Burdette, PhD, University of Illinois at Chicago, 900 S. Ashland Ave, Chicago, IL 60607, USA.
| |
Collapse
|
27
|
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
|
28
|
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
|
29
|
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
|
30
|
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
|
31
|
He Y, Meng K, Wang X, Dong Z, Zhang Y, Quan F. Comparison of Bovine Small Antral Follicle Development in Two- and Three-Dimensional Culture Systems. AN ACAD BRAS CIENC 2020; 92:e20180935. [PMID: 33146258 DOI: 10.1590/0001-3765202020180935] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 12/13/2018] [Indexed: 12/26/2022] Open
Abstract
To compare the effects of two-(2D, microplate) and three-dimensional (3D, alginate) culture systems on the in vitro growth of small antral follicles in cattle, individual follicles were separately cultured in the two culture systems for 8 days. Half of the culture medium was replaced by fresh medium every 2 days; the former medium was used to assess the amount of follicular hormone secretion using ELISA. Individual follicle morphology, diameter, and survival rate were recorded every alternate day. The results showed that in 4 days, there was no significant difference between the two systems, except that the growth rate of follicles in 2D system was relatively faster. After 4 days, estradiol concentration in 3D system was higher than that in 2D system. However, progesterone concentration was lower than that in the 2D system. The survival rate and oocyte quality of follicles in 2D system were significantly lower than those in 3D system on day 8. The follicle diameter slightly increased (30-60 μm) in the entire process. Taken together, for in vitro culture of follicles within 4 days, the 2D culture system is more suitable. However, when the culture duration is >4 days, the 3D culture system is more suitable.
Collapse
Affiliation(s)
- Yuanyuan He
- Northwest A&F University, College of Veterinary Medicine, Department of Clinical Veterinary Medicine, Yangling 712100 Shaanxi, China
| | - Kai Meng
- Northwest A&F University, College of Veterinary Medicine, Department of Clinical Veterinary Medicine, Yangling 712100 Shaanxi, China
| | - Xiaomei Wang
- Northwest A&F University, College of Veterinary Medicine, Department of Clinical Veterinary Medicine, Yangling 712100 Shaanxi, China
| | - Zhihang Dong
- Northwest A&F University, College of Veterinary Medicine, Department of Clinical Veterinary Medicine, Yangling 712100 Shaanxi, China
| | - Yong Zhang
- Northwest A&F University, College of Veterinary Medicine, Department of Clinical Veterinary Medicine, Yangling 712100 Shaanxi, China
| | - Fusheng Quan
- Northwest A&F University, College of Veterinary Medicine, Department of Clinical Veterinary Medicine, Yangling 712100 Shaanxi, China
| |
Collapse
|
32
|
Yoon JD, Hwang SU, Kim M, Jeon Y, Hyun SH. Growth differentiation factor 8 regulates SMAD2/3 signaling and improves oocyte quality during porcine oocyte maturation in vitro†. Biol Reprod 2020; 101:63-75. [PMID: 31004472 DOI: 10.1093/biolre/ioz066] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 08/07/2018] [Accepted: 04/17/2019] [Indexed: 11/12/2022] Open
Abstract
Growth differentiation factor 8 (GDF8), also known as myostatin, is a member of the transforming growth factor-β (TGF-β) family and has been identified as a strong physiological regulator of muscle differentiation. Recently, the functional role of GDF8 in reproductive organs has received increased interest following its detection in the human placenta and uterus. To investigate the effects of GDF8 during porcine oocyte in vitro maturation (IVM), we assessed the quality of matured oocytes. Furthermore, we investigated the specific gene transcription and protein activation levels in oocytes and cumulus cells after IVM and subsequent embryonic development after in vitro fertilization and parthenogenetic activation. Prior to these experiments, the concentration of GDF8 in porcine follicular fluid was determined. During the entire IVM period, 1.3 ng/mL GDF8 and its signaling inhibitor SB431542 (SB) at 5 μM were added as control, SB, SB + GDF8, and GDF8 groups, respectively. Our results demonstrate that supplementation with GDF8 during porcine oocyte IVM enhanced both meiotic and cytoplasmic maturation, with altered transcriptional patterns, via activation of Sma- and Mad-related protein 2/3 (SMAD2/3). Using the pharmacological inhibitor SB431542, we demonstrated that inhibition of GDF8-induced Smad2/3 signaling reduces matured oocyte quality. In conclusion, for the first time, we demonstrated paracrine factor GDF8 in porcine follicular fluid in vivo. Furthermore, we showed that GDF8 supplementation improved mature oocyte quality by regulating p38 mitogen-activated protein kinase phosphorylation and intracellular glutathione and reactive oxygen species levels during porcine IVM.
Collapse
Affiliation(s)
- Junchul David Yoon
- Laboratory of Veterinary Embryology and Biotechnology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea.,Institute of Stem Cell & Regenerative Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Seon-Ung Hwang
- Laboratory of Veterinary Embryology and Biotechnology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea.,Institute of Stem Cell & Regenerative Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Mirae Kim
- Laboratory of Veterinary Embryology and Biotechnology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea.,Institute of Stem Cell & Regenerative Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Yubyeol Jeon
- Laboratory of Theriogenology and Reproductive Biotechnologies, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeolabuk-do, Republic of Korea
| | - Sang-Hwan Hyun
- Laboratory of Veterinary Embryology and Biotechnology, Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea.,Institute of Stem Cell & Regenerative Medicine, Chungbuk National University, Cheongju, Republic of Korea
| |
Collapse
|
33
|
Zubizarreta ME, Xiao S. Bioengineering models of female reproduction. Biodes Manuf 2020; 3:237-251. [PMID: 32774987 PMCID: PMC7413245 DOI: 10.1007/s42242-020-00082-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 05/23/2020] [Indexed: 12/25/2022]
Abstract
The female reproductive system consists of the ovaries, the female gonads, and the reproductive track organs of the fallopian tubes, uterus, cervix, and vagina. It functions to provide hormonal support and anatomical structure for the production of new offspring. A number of endogenous and exogenous factors can impact female reproductive health and fertility, including genetic vulnerability, medications, environmental exposures, age, nutrition, and diseases, etc. To date, due to the ethical concerns of using human subjects in biomedical research, the majority of studies use in vivo animal models and 2D cell/tissue culture models to study female reproduction. However, the complexity and species difference of the female reproductive system in humans makes it difficult to compare to those of animals. Moreover, the monolayered cells cultured on flat plastics or glass lose their 3D architecture as well as the physical and/or biochemical contacts with other cells in vivo. Further, all reproductive organs do not work alone but interconnect with each other and also with non-reproductive organs to support female reproductive, endocrine, and systemic health. These facts suggest that there is an urgent and unmet need to develop representative, effective, and efficient in vitro models for studying human female reproduction. The prodigious advancements of bioengineering (e.g. biomaterials, 3D printing, and organ-on-a-chip) allow us to study female reproduction in an entirely new way. Here, we review recent advances that use bioengineering methods to study female reproduction, including the bioengineering models of the ovary, fallopian tube, uterus, embryo implantation, placenta, and reproductive disease.
Collapse
Affiliation(s)
- Maria E. Zubizarreta
- Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Shuo Xiao
- Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Environmental Health Sciences Institute, Rutgers University, Piscataway, NJ, 08854, USA
| |
Collapse
|
34
|
Jalili C, Khani Hemmatabadi F, Mansouri K, Bakhtiyari M. Effects of sodium alginate capsules as 3D scaffolds on hormones and genes expression in preantral follicles of mice compared to 2D medium: An experimental study. Int J Reprod Biomed 2020; 18:517-530. [PMID: 32803116 PMCID: PMC7385913 DOI: 10.18502/ijrm.v13i7.7369] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 06/18/2019] [Accepted: 01/14/2020] [Indexed: 12/02/2022] Open
Abstract
Background The improvement of in vitro maturation methods, which can activate the preantral follicle growth, plays a crucial role in the production of mature oocytes in reproductive technology. Objective To evaluate the different concentrations of 3D scaffolds of sodium alginate on hormones and gene expression in mice preantral follicles. Materials and Methods Immature female BALB/c mice (12-14 days) were sacrificed. The follicles were removed mechanically and transferred into α minimal essential medium with 5% fetal bovine serum. The preantral follicles were incubated with different concentrations of sodium alginate (0.25%, 0.5%, and 1%) and 2D medium for 12 days. The follicles were examined for antral formation following the 10th day and the diameter on days 6 th and 12 th . The levels of hormones (AMH, androstenedione, 17β-estradiol, and progesterone) and the expression of genes (CYP11a1, CYP17a1, CYP19a1, AMH, and GnRH) at the end of the 12 th day. Results Maximum follicle diameter and highest percentage of antrum formation were related to 0.5% concentration (p = 0.00). The levels of hormones in different doses of sodium alginate were increased significantly compared to the control group (p = 0.00). The highest and lowest levels of these hormones were related to 0.5% concentration and 2D medium, respectively. The highest level of genes expression was observed in 0.5% sodium alginate, which showed a significant increase compared to the control group (p = 0.00). Conclusion Proper concentration of alginate hydrogel increases follicle growth, causes follicle maturation, produces steroid hormones, and increases appropriate expression of steroidogenesis-related genes.
Collapse
Affiliation(s)
- Cyrus Jalili
- Department of Anatomical Sciences, Medical Biology Research Center, Kermanshah University of Medical Sciences, Taghbostan, Kermanshah, Iran
| | | | - Kamran Mansouri
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mehrdad Bakhtiyari
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
35
|
Woodruff TK. Lessons from bioengineering the ovarian follicle: a personal perspective. Reproduction 2020; 158:F113-F126. [PMID: 31846436 DOI: 10.1530/rep-19-0190] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 07/15/2019] [Indexed: 12/14/2022]
Abstract
The ovarian follicle and its maturation captivated my imagination and inspired my scientific journey - what we know now about this remarkable structure is captured in this invited review. In the past decade, our knowledge of the ovarian follicle expanded dramatically as cross-disciplinary collaborations brought new perspectives to bear, ultimately leading to the development of extragonadal follicles as model systems with significant clinical implications. Follicle maturation in vitro in an 'artificial' ovary became possible by learning what the follicle is fundamentally and autonomously capable of - which turns out to be quite a lot. Progress in understanding and harnessing follicle biology has been aided by engineers and materials scientists who created hardware that enables tissue function for extended periods of time. The EVATAR system supports extracorporeal ovarian function in an engineered environment that mimics the endocrine environment of the reproductive tract. Finally, applying the tools of inorganic chemistry, we discovered that oocytes require zinc to mature over time - a truly new aspect of follicle biology with no antecedent other than the presence of zinc in sperm. Drawing on the tools and ideas from the fields of bioengineering, materials science and chemistry unlocked follicle biology in ways that we could not have known or even predicted. Similarly, how today's basic science discoveries regarding ovarian follicle maturation are translated to improve the experience of tomorrow's patients is yet to be determined.
Collapse
Affiliation(s)
- Teresa K Woodruff
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
36
|
Simon LE, Kumar TR, Duncan FE. In vitro ovarian follicle growth: a comprehensive analysis of key protocol variables†. Biol Reprod 2020; 103:455-470. [PMID: 32406908 DOI: 10.1093/biolre/ioaa073] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 05/06/2020] [Accepted: 05/12/2020] [Indexed: 12/18/2022] Open
Abstract
Folliculogenesis is a complex process that requires integration of autocrine, paracrine, and endocrine factors together with tightly regulated interactions between granulosa cells and oocytes for the growth and survival of healthy follicles. Culture of ovarian follicles is a powerful approach for investigating folliculogenesis and oogenesis in a tightly controlled environment. This method has not only enabled unprecedented insight into the fundamental biology of follicle development but also has far-reaching translational applications, including in fertility preservation for women whose ovarian follicles may be damaged by disease or its treatment or in wildlife conservation. Two- and three-dimensional follicle culture systems have been developed and are rapidly evolving. It is clear from a review of the literature on isolated follicle culture methods published over the past two decades (1980-2018) that protocols vary with respect to species examined, follicle isolation methods, culture techniques, culture media and nutrient and hormone supplementation, and experimental endpoints. Here we review the heterogeneity among these major variables of follicle culture protocols.
Collapse
Affiliation(s)
- Leah E Simon
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - T Rajendra Kumar
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA.,Department of Obstetrics and Gynecology, University of Colorado, Aurora, Colorado, USA
| | - Francesca E Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
37
|
The new biocompatible material for mouse ovarian follicle development in three-dimensional in vitro culture systems. Theriogenology 2020; 144:33-40. [DOI: 10.1016/j.theriogenology.2019.12.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/15/2019] [Accepted: 12/15/2019] [Indexed: 12/30/2022]
|
38
|
Candelaria JI, Denicol AC. Characterization of isolated bovine preantral follicles based on morphology, diameter and cell number. ZYGOTE 2020; 28:1-6. [PMID: 31933452 DOI: 10.1017/s0967199419000832] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Preantral follicles are a potential reservoir of oocytes to be used in assisted reproductive technologies. With the increasing interest in developing techniques to grow preantral follicles in vitro, and as the bovine emerges as an appropriate model species to understand human folliculogenesis, the establishment of an accurate classification of developmental stages is needed. Classification of bovine preantral follicles has been mostly based on histological analysis and estimation models, which may not translate well to correctly characterize preantral follicles isolated from the ovary. In this study, we classified bovine preantral follicles by morphology upon isolation, determined diameter and number of granulosa cells by direct counting, and compared our results with previous studies reporting bovine preantral follicle classification. Follicles were isolated via homogenization of ovary tissue and classified into primary, early secondary and secondary stage based on morphology and number of layers of granulosa cells. Diameter was individually measured and Hoechst 33342 was used as a nuclear stain to count granulosa cells. We found that follicles classified by morphology into primary, early secondary, and secondary had different mean diameter and cell number (P < 0.01); cell number and diameter were positively correlated, as were cell density and cell number in each developmental stage (P < 0.01). Results obtained here were mostly in agreement with previous classifications based on histological sections and on isolated follicles, with some discrepancies. The present data add accuracy to classification of bovine preantral follicles that is critical to optimize culture conditions to produce developmentally competent oocytes.
Collapse
Affiliation(s)
| | - Anna C Denicol
- Department of Animal Science, University of California, Davis, CA, USA
| |
Collapse
|
39
|
Barros VRP, Monte APO, Lins TLBG, Santos JM, Menezes VG, Cavalcante AYP, Araújo VR, Gouveia BB, Matos MHT. In vitro survival, growth, and maturation of sheep oocytes from secondary follicles cultured in serum-free conditions: impact of a constant or a sequential medium containing recombinant human FSH. Domest Anim Endocrinol 2019; 67:71-79. [PMID: 30765293 DOI: 10.1016/j.domaniend.2018.12.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 12/07/2018] [Accepted: 12/12/2018] [Indexed: 12/16/2022]
Abstract
This study evaluated the in vitro development and maturation of ovine oocytes from secondary follicles cultured in serum-free medium containing fixed or sequential concentrations of recombinant human FSH (rhFSH). Follicles were cultured in α-MEM+ alone or with constant (500, 750, or 1,000 ng/mL) or sequential concentrations of rhFSH (seq. 1: day 6 = 500; day 12 = 750; day 18 = 1,000 ng/mL and seq. 2: day 6 = 100; day 12 = 500; day 18 = 1,000 ng/mL). At the end of the experiment, follicular survival was higher (P < 0.05) in 750 ng/mL rhFSH than the control and 1,000 ng/mL rhFSH. As early as day 6 of culture, antral cavity formation was observed in all treatments. Follicular diameter increased progressively and significantly in all treatments throughout 18 d of culture. Furthermore, addition of rhFSH to the medium promoted a significant increase in the percentage of fully grown oocytes in all treatments compared to α-MEM+. Mitochondrial activity was higher in rhFSH treatments than in the control, except in rhFSH seq. 2 (P < 0.05). Maturation rates increased in oocytes from intact follicles cultured in 750 ng/mL rhFSH compared to the control (P < 0.05). In conclusion, rhFSH at 750 ng/mL maintained the survival of secondary follicles cultured in serum-free medium, improved oocyte growth, mitochondrial activity, and oocyte maturation.
Collapse
Affiliation(s)
- V R P Barros
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Petrolina, PE 56300-990, Brazil
| | - A P O Monte
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Petrolina, PE 56300-990, Brazil
| | - T L B G Lins
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Petrolina, PE 56300-990, Brazil
| | - J M Santos
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Petrolina, PE 56300-990, Brazil
| | - V G Menezes
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Petrolina, PE 56300-990, Brazil
| | - A Y P Cavalcante
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Petrolina, PE 56300-990, Brazil
| | - V R Araújo
- Health Center Science, State University of Ceará, 60714-903 Fortaleza, CE, Brazil
| | - B B Gouveia
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Petrolina, PE 56300-990, Brazil
| | - M H T Matos
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Federal University of São Francisco Valley, Petrolina, PE 56300-990, Brazil.
| |
Collapse
|
40
|
Equine chorionic gonadotropin induces in vitro follicular growth from the multi-layered secondary developmental stage in cats. Theriogenology 2019; 123:116-122. [DOI: 10.1016/j.theriogenology.2018.09.040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 09/27/2018] [Accepted: 09/27/2018] [Indexed: 12/16/2022]
|
41
|
Influence of follicle-stimulating hormone concentrations on the integrity and development of bovine follicles cultured in vitro. ZYGOTE 2018; 26:417-423. [DOI: 10.1017/s0967199418000497] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
SummaryThis study investigated the in vitro culture of bovine follicles included in ovarian tissue for 2 or 6 days (D2 or D6), with the addition of different concentrations of follicle-stimulating hormone (FSH) (0, 10, 50, 100 or 200 ng/ml). Data were compared for follicular development, morphological integrity and diameter of follicles and oocytes. Ovaries (n = 10) from Nelore cows (n = 5) were divided into fragments (n = 11 per ovary) and were immediately fixed in Bouin’s solution (D0) or were individually cultured for 2 or 6 days in one of the described concentrations of FSH and then processed for histology. Compared with the rates of follicular development at D2 for minimal essential medium (MEM) (75.0%) and 50 ng/ml of FSH (71.1%), the best rates of follicular development at D2 were obtained with 10 (84.7%), 100 (87.5%) and 200 ng/ml of FSH (85.0%; P<0.05). After 6 days of cultivation, there were no differences among treatments regarding follicular growth. The morphological integrity of preantral follicles was better maintained by 100 ng/ml FSH for 2 and 6 days of cultivation (51.2 and 40.4%, respectively; P<0.05) than that for MEM (D2: 30.9%, D6: 20.8%), 10 (D2: 39.2%, D6: 22.8%), 50 (D2: 30.4%, D6: 28.8%) and 200 ng/ml FSH (D2: 45.2%, D6: 36.8%). FSH at 100 ng/ml provided the highest mean diameter averages: 34.5±10.8 µm at D2 and 33.2±12.5 µm at D6 (P<0.05). We concluded that the medium supplemented with 100 ng/ml FSH during in vitro culture provided appropriate conditions for the development and morphological integrity of preantral follicles in cattle.
Collapse
|
42
|
Baba T, Ting AY, Tkachenko O, Xu J, Stouffer RL. Direct actions of androgen, estrogen and anti-Müllerian hormone on primate secondary follicle development in the absence of FSH in vitro. Hum Reprod 2018; 32:2456-2464. [PMID: 29077845 DOI: 10.1093/humrep/dex322] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 10/03/2017] [Indexed: 11/14/2022] Open
Abstract
STUDY QUESTION What are effects of androgen, estrogen and anti-Müllerian hormone (AMH), independent of FSH action, on the development and function of primate follicles from the preantral to small antral stage in vitro? SUMMARY ANSWER Androgen and estrogen, but not AMH, promote follicle survival and growth in vitro, in the absence of FSH. However, their growth-promoting effects are limited to the preantral to early antral stage. WHAT IS KNOWN ALREADY FSH supports primate preantral follicle development in vitro. Androgen and estrogen augment follicle survival and growth in the presence of FSH during culture. STUDY DESIGN SIZE, DURATION Nonhuman primate model; randomized, control versus treatment groups. Rhesus macaque (n = 6) secondary follicles (n = 24 per animal per treatment group) were cultured for 5 weeks. PARTICIPANTS/MATERIALS, SETTING, METHODS Follicles were encapsulated in 0.25% (w/v) alginate and cultured individually in modified alpha minimum essential media with (i) FSH (1 ng/ml; control), (ii) no FSH, (iii) no FSH + estradiol (E2; 100 pg/ml)/dihydrotestosterone (DHT; 50 ng/ml) and (iv) no FSH + AMH (50 ng/ml). In a second experiment, follicles were cultured with (i) FSH (1 ng/ml), (ii) no FSH, (iii) no FSH + E2 (1 ng/ml), (iv) no FSH + DHT (50 ng/ml) and (v) no FSH + E2/DHT. Follicle survival, antrum formation and growth pattern were evaluated. Progesterone (P4), E2 and AMH concentrations in culture media were measured. MAIN RESULTS AND THE ROLE OF CHANCE In the first experiment, FSH deprivation significantly decreased (P < 0.05) follicle survival rates in the no FSH group (16 ± 5%), compared to CTRL (66 ± 9%). E2/DHT (49 ± 5%), but not AMH (27 ± 8%), restored follicle survival rate to the CTRL level. Similarly, antrum formation rates were higher (P < 0.05) in CTRL (56 ± 6%) and E2/DHT groups (54 ± 14%), compared to no FSH (0 ± 0%) and AMH (11 ± 11%) groups. However, follicle growth rate after antrum formation and follicle diameter at week 5 was reduced (P < 0.05) in the E2/DHT group (405 ± 25 μm), compared to CTRL (522 ± 29 μm). Indeed, the proportion of fast-grow follicles at week 5 was higher in CTRL (29% ± 5), compared to E2/DHT group (10 ± 3%). No fast-grow follicles were observed in no FSH and AMH groups. AMH levels at week 3 remained similar in all groups. However, media concentrations of P4 and E2 at week 5 were lower (P < 0.05, undetectable) in no FSH, E2/DHT and AMH groups, compared to CTRL (P4 = 93 ± 10 ng/ml; E2 = 4 ± 1 ng/ml). In the second experiment, FSH depletion diminished follicle survival rate (66 ± 8% in control versus 45 ± 9% in no FSH, P = 0.034). E2 plus DHT (31.5 ± 11%) or DHT alone (69 ± 9%) restored follicle survival rate to the control (FSH) level as expected. Also, E2 plus DHT or DHT alone improved antrum formation rate. However, in the absence of FSH, E2 plus DHT or DHT alone did not support growth, in terms of follicle diameter, or steroid (P4 or E2) production after the antral stage. LIMITATIONS REASONS FOR CAUTION This study is limited to in vitro effects of E2, DHT and AMH during the interval from the secondary to small antral stage of macaque follicular development. In addition, the primate follicle pool is heterogeneous and differs between animals; therefore, even though only secondary follicles were selected, follicle growth and developmental outcomes might differ from one animal to another. WIDER IMPLICATIONS OF THE FINDINGS This study provides novel information on the possible actions of estrogen and androgen during early follicular development in primates. Our results suggest that sequential exposure of preantral follicles to local factors, e.g. E2 and DHT, followed by gonadotropin once the follicle reaches the antral stage, may better mimic primate folliculogenesis in vivo. STUDY FUNDING/COMPETING INTEREST(S) Research reported in this publication was supported by the Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Center for Translational Research on Reproduction and Infertility 5P50HD071836, and the NIH Primate Centers Program 8P510D011092. There are no conflicts of interest.
Collapse
Affiliation(s)
- T Baba
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, 505 NW 185th Avenue, Beaverton, OR 97006, USA
- Department of Obstetrics and Gynecology, Sapporo Medical University, South 1 West 16, Sapporo, Hokkaido 060-8543 Japan
| | - A Y Ting
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, 505 NW 185th Avenue, Beaverton, OR 97006, USA
| | - O Tkachenko
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, 505 NW 185th Avenue, Beaverton, OR 97006, USA
| | - J Xu
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, 505 NW 185th Avenue, Beaverton, OR 97006, USA
- Department of Obstetrics and Gynecology, School of Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - R L Stouffer
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, 505 NW 185th Avenue, Beaverton, OR 97006, USA
- Department of Obstetrics and Gynecology, School of Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| |
Collapse
|
43
|
Leal ÉSS, Vieira LA, Sá NAR, Silva GM, Lunardi FO, Ferreira ACA, Campello CC, Alves BG, Cibin FWS, Smitz J, Figueiredo JR, Rodrigues APR. In vitro growth and development of isolated secondary follicles from vitrified caprine ovarian cortex. Reprod Fertil Dev 2018; 30:359-370. [PMID: 28768567 DOI: 10.1071/rd16487] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Accepted: 06/17/2017] [Indexed: 12/27/2022] Open
Abstract
The aim of this study was to evaluate the viability, antrum formation and in vitro development of isolated secondary follicles from vitrified caprine ovarian cortex in a medium previously established for fresh isolated secondary follicles, in the absence (α-minimum essential medium (α-MEM+) alone) or presence of FSH and vascular endothelial growth factor (VEGF; α-MEM++FSH+VEGF). Ovarian fragments were distributed among five treatments (T1 to T5): fresh follicles were fixed immediately (T1), follicles from fresh tissue were cultured in vitro in α-MEM+ (T2) or α-MEM++FSH+VEGF (T3) and follicles from vitrified tissue were cultured in vitro in α-MEM+ (T4) or α-MEM++FSH+VEGF (T5). After 6 days of culture, treated follicles (T2, T3, T4 and T5) were evaluated for morphology, viability and follicular development (growth, antrum formation and proliferation of granulosa cells by Ki67 and argyrophilic nucleolar organiser region (AgNOR) staining). The levels of reactive oxygen species (ROS) in the culture media were also assessed. Overall, morphology of vitrified follicles was altered (P<0.05) compared with the fresh follicles. Follicular viability, antrum formation and ROS were similar between treatments (P>0.05). The average overall and daily follicular growth was highest (P<0.05) in T3. Granulosa cells in all treatments (T1, T2, T3, T4 and T5) stained positive for Ki67. However, fresh follicles from T3 had significantly higher AgNOR staining (P<0.05) compared with follicles of T1, T2, T4 and T5. In conclusion, secondary follicles can be isolated from vitrified and warmed ovarian cortex and survive and form an antrum when growing in an in vitro culture for 6 days.
Collapse
Affiliation(s)
- Érica S S Leal
- Faculty of Veterinary Medicine, LAMOFOPA, PPGCV, State University of Ceará, Av. Dr Silas Munguba, 1700 - Campus of Itaperi, Fortaleza - CE - CEP 60741-903, Brazil
| | - Luis A Vieira
- Faculty of Veterinary Medicine, LAMOFOPA, PPGCV, State University of Ceará, Av. Dr Silas Munguba, 1700 - Campus of Itaperi, Fortaleza - CE - CEP 60741-903, Brazil
| | - Naíza A R Sá
- Faculty of Veterinary Medicine, LAMOFOPA, PPGCV, State University of Ceará, Av. Dr Silas Munguba, 1700 - Campus of Itaperi, Fortaleza - CE - CEP 60741-903, Brazil
| | - Gerlane M Silva
- Faculty of Veterinary Medicine, LAMOFOPA, PPGCV, State University of Ceará, Av. Dr Silas Munguba, 1700 - Campus of Itaperi, Fortaleza - CE - CEP 60741-903, Brazil
| | - Franciele O Lunardi
- Faculty of Veterinary Medicine, LAMOFOPA, PPGCV, State University of Ceará, Av. Dr Silas Munguba, 1700 - Campus of Itaperi, Fortaleza - CE - CEP 60741-903, Brazil
| | - Anna C A Ferreira
- Faculty of Veterinary Medicine, LAMOFOPA, PPGCV, State University of Ceará, Av. Dr Silas Munguba, 1700 - Campus of Itaperi, Fortaleza - CE - CEP 60741-903, Brazil
| | - Cláudio C Campello
- Faculty of Veterinary Medicine, LAMOFOPA, PPGCV, State University of Ceará, Av. Dr Silas Munguba, 1700 - Campus of Itaperi, Fortaleza - CE - CEP 60741-903, Brazil
| | - Benner G Alves
- Faculty of Veterinary Medicine, LAMOFOPA, PPGCV, State University of Ceará, Av. Dr Silas Munguba, 1700 - Campus of Itaperi, Fortaleza - CE - CEP 60741-903, Brazil
| | - Francielli W S Cibin
- University Federal of Pampa, Uruguaiana-Rio Grande do Sul, Av. General Osório, 900 - São Jorge Bagé, RS - CE - 96400-100, Brazil
| | - Johan Smitz
- Follicle Biology Laboratory, Center for Reproductive Medicine, UZ Brussel, Laarbeeklaan 101, B-1090 Brussels, Belgium
| | - José R Figueiredo
- Faculty of Veterinary Medicine, LAMOFOPA, PPGCV, State University of Ceará, Av. Dr Silas Munguba, 1700 - Campus of Itaperi, Fortaleza - CE - CEP 60741-903, Brazil
| | - Ana P R Rodrigues
- Faculty of Veterinary Medicine, LAMOFOPA, PPGCV, State University of Ceará, Av. Dr Silas Munguba, 1700 - Campus of Itaperi, Fortaleza - CE - CEP 60741-903, Brazil
| |
Collapse
|
44
|
Synergy of Paracrine Signaling During Early-Stage Mouse Ovarian Follicle Development In Vitro. Cell Mol Bioeng 2018; 11:435-450. [PMID: 31719893 DOI: 10.1007/s12195-018-0545-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 07/20/2018] [Indexed: 10/28/2022] Open
Abstract
Introduction Paracrine signals, such as soluble cytokines and extracellular matrix cues, are essential for the survival and development of multicellular ovarian follicles. While it is well established that hydrogel-based culture systems successfully support the growth of late-stage follicles for fertility preservation, growing small, early-stage ovarian follicles still proves to be challenging. We hypothesized that paracrine factors secreted from neighboring follicles may be crucial for improving the survival of early-stage follicles in vitro. Methods To test our hypothesis, we investigated the bi-directional crosstalk of the paracrine signals, such as cell-secreted cytokines, sex hormones and transcription factors (TFs), in follicles encapsulated and cultured for 12 days in alginate in groups of five (5×) and ten (10×). Results The differential profiles of TF activity and secretome during folliculogenesis were analyzed using TRanscriptional Activity CEllular aRray (TRACER) and data-driven multivariate modeling approach. The mechano- and oxygen-responsive TFs, NF-κB and HIF1, exhibited a unique upregulation signature in 10× follicles. Consistently, levels of proangiogenic factors, such as VEGF-A and angiopoietin-2, were significantly higher in 10× follicles than those in 5× follicles, reaching 269.77 and 242.82 pg/mL on the last day of culture. The analysis of TRACER and secreted cytokines also revealed critical early interactions between cytokines and TFs, correlating with the observed phenotypical and functional differences between conditions. Conclusions We identified unique signatures of synergism during successful early-stage ovarian follicle development. These findings bring us closer to understanding of mechanisms underlying the downstream effects of interactions between the extracellular microenvironment and early-stage folliculogenesis in vitro.
Collapse
|
45
|
Fabbri R, Zamboni C, Vicenti R, Macciocca M, Paradisi R, Seracchioli R. Update on oogenesis in vitro. ACTA ACUST UNITED AC 2018; 70:588-608. [PMID: 29999288 DOI: 10.23736/s0026-4784.18.04273-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Ovarian tissue is increasingly being collected from cancer patients and cryopreserved for fertility preservation. Alternately to the autologous transplantation, the development of culture systems that support oocyte development from the primordial follicle stage represent a valid strategy to restore fertility. The aim of this study is to review the most recent data regarding oogenesis in vitro and to provide an up-to-date on the contemporary knowledge of follicle growth and development in vitro. EVIDENCE ACQUISITION A comprehensive systematic MEDLINE search was performed since February 2018 for English-language reports by using the following terms: "ovary," "animal and human follicle," "in vitro growth and development," "ovarian tissue culture," "fertility preservation," "IVM," "oocyte." Previous published reviews and recent published original articles were preferred in order to meet our study scope. EVIDENCE SYNTHESIS Over time, many studies have been conducted with the aim to optimize the characteristics of ovarian tissue culture systems and to better support the three main phases: 1) activation of primordial follicles; 2) isolation and culture of growing preantral follicles; 3) removal from the follicle environment and maturation of oocyte cumulus complexes. While complete oocyte in vitro development has been achieved in mouse, with the production of live offspring, the goal of obtaining oocytes of sufficient quality to support embryo development has not been completely reached into higher mammals despite decades of effort. CONCLUSIONS Over the years, many improvements have been made on ovarian tissue cultures with the future purpose that patients will be provided with a greater number of developmentally competent oocytes for fertility preservation.
Collapse
Affiliation(s)
- Raffaella Fabbri
- Unit of Gynecology and Physiopathology of Human Reproduction, Department of Medical and Surgical Sciences, University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Chiara Zamboni
- Unit of Gynecology and Physiopathology of Human Reproduction, Department of Medical and Surgical Sciences, University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy -
| | - Rossella Vicenti
- Unit of Gynecology and Physiopathology of Human Reproduction, Department of Medical and Surgical Sciences, University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Maria Macciocca
- Unit of Gynecology and Physiopathology of Human Reproduction, Department of Medical and Surgical Sciences, University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| | | | - Renato Seracchioli
- Unit of Gynecology and Physiopathology of Human Reproduction, Department of Medical and Surgical Sciences, University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| |
Collapse
|
46
|
Nagashima JB, El Assal R, Songsasen N, Demirci U. Evaluation of an ovary-on-a-chip in large mammalian models: Species specificity and influence of follicle isolation status. J Tissue Eng Regen Med 2018; 12:e1926-e1935. [PMID: 29222841 DOI: 10.1002/term.2623] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 11/09/2017] [Accepted: 11/27/2017] [Indexed: 12/20/2022]
Abstract
The ability to grow oocytes from immature ovarian follicles in vitro has significant potential for fertility preservation; yet, it has proved challenging in large mammalian species due to the complex metabolic needs and long-term culture requirements. Currently, follicular incubations are based on a "static" system with manual exchange of medium. Despite the numerous advantages of conventional culturing approaches, recapitulating the native microenvironment and supporting the survival of ovarian follicles from large mammalian species still represent challenges. In this study, we utilized an innovative, dynamic microfluidic system to support the in vitro survival of domestic cat and dog follicles enclosed within the ovarian cortex or isolated from ovarian cortex. Results indicate both species-specific and tissue type-specific differences in response to microfluidic culture. Domestic cat but not dog ovarian cortical tissues maintained viability under flow similar to conventional agarose gel controls. Preantral stage isolated follicles from both species that grew most favourably in conventional alginate bead culture, but overall, there was no influence of culture system on expression of follicle development or oocyte health markers. This system represents an important exploration toward the development of an improved ovarian in vitro culture system of large mammalian species (e.g., cats and dogs), which has potential applications for fertility preservation, reproductive toxicology, and endangered mammal conservation efforts.
Collapse
Affiliation(s)
- Jennifer B Nagashima
- Smithsonian Conservation Biology Institute, National Zoological Park, Front Royal, VA, USA.,Bio-Acoustic-MEMS in Medicine (BAMM) Laboratories, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Rami El Assal
- Bio-Acoustic-MEMS in Medicine (BAMM) Laboratories, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Nucharin Songsasen
- Smithsonian Conservation Biology Institute, National Zoological Park, Front Royal, VA, USA
| | - Utkan Demirci
- Bio-Acoustic-MEMS in Medicine (BAMM) Laboratories, Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA, USA.,Department of Electrical Engineering (by courtesy), Stanford University School of Engineering, Palo Alto, CA, USA
| |
Collapse
|
47
|
Richards JS, Ren YA, Candelaria N, Adams JE, Rajkovic A. Ovarian Follicular Theca Cell Recruitment, Differentiation, and Impact on Fertility: 2017 Update. Endocr Rev 2018; 39:1-20. [PMID: 29028960 PMCID: PMC5807095 DOI: 10.1210/er.2017-00164] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 09/12/2017] [Indexed: 12/24/2022]
Abstract
The major goal of this review is to summarize recent exciting findings that have been published within the past 10 years that, to our knowledge, have not been presented in detail in previous reviews and that may impact altered follicular development in polycystic ovarian syndrome (PCOS) and premature ovarian failure in women. Specifically, we will cover the following: (1) mouse models that have led to discovery of the derivation of two precursor populations of theca cells in the embryonic gonad; (2) the key roles of the oocyte-derived factor growth differentiation factor 9 on the hedgehog (HH) signaling pathway and theca cell functions; and (3) the impact of the HH pathway on both the specification of theca endocrine cells and theca fibroblast and smooth muscle cells in developing follicles. We will also discuss the following: (1) other signaling pathways that impact the differentiation of theca cells, not only luteinizing hormone but also insulinlike 3, bone morphogenic proteins, the circadian clock genes, androgens, and estrogens; and (2) theca-associated vascular, immune, and fibroblast cells, as well as the cytokines and matrix factors that play key roles in follicle growth. Lastly, we will integrate what is known about theca cells from mouse models, human-derived theca cell lines from patients who have PCOS and patients who do not have PCOS, and microarray analyses of human and bovine theca to understand what pathways and factors contribute to follicle growth as well as to the abnormal function of theca.
Collapse
Affiliation(s)
- JoAnne S. Richards
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Yi A. Ren
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Nicholes Candelaria
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Jaye E. Adams
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Aleksandar Rajkovic
- Department of Obstetrics, Gynecology and Reproductive Medicine, Magee-Women’s Research Institute, Pittsburgh, Pennsylvania 15213
| |
Collapse
|
48
|
Sadr SZ, Fatehi R, Maroufizadeh S, Amorim CA, Ebrahimi B. Utilizing Fibrin-Alginate and Matrigel-Alginate for Mouse Follicle Development in Three-Dimensional Culture Systems. Biopreserv Biobank 2018; 16:120-127. [PMID: 29363997 DOI: 10.1089/bio.2017.0087] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In vitro culture of ovarian follicles is a new technique in reproductive technology, which helps in understanding the process of folliculogenesis. The in vitro culture of follicles could be carried out using three-dimensional (3D) natural scaffolds that mimic the ovarian tissue stroma. Selection of the right matrix and culture media in these scaffolds could increase the survival and maturation of the follicles. In this work, the applicability of matrigel-alginate (MA) and fibrin-alginate (FA) 3D scaffolds for folliculogenesis was assessed. The ovaries of 13-day-old Naval Medical Research Institute (NMRI) mice were isolated and distributed into control and vitrification groups. Preantral follicles (mean diameter: 120-140 μm) were mechanically isolated from control and vitrified-warmed ovaries, encapsulated in MA or FA scaffold and cultured for 12 days. Follicle survival, growth, maturation, and quantitative expression of oocyte maturation genes (Gdf9, Bmp15, Fgf8, KitL, Kit, and Amh) and proteins (GDF9 and BMP15) were assessed. Survival rate of culture preantral follicles in control groups was found to be significantly higher than vitrified follicles. Antrum formation was similar in all groups. Follicle diameters were significantly increased in all groups during culture period. A decreasing pattern of gene expression was seen for all genes in all groups. This trend was verified through evaluation of protein expression, during which there was strong staining in antral follicles from all groups in the last day of in vitro culture. The better survival and maturation rate of follicles in the MA compared to FA scaffold indicates that the MA matrix, being rich in extracellular matrix components, could mimic the ovarian condition better and presents a good environment for follicle development.
Collapse
Affiliation(s)
- Seyedeh Zeynab Sadr
- 1 Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine , ACECR, Tehran, Iran
| | - Roya Fatehi
- 1 Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine , ACECR, Tehran, Iran
| | - Saman Maroufizadeh
- 2 Department of Epidemiology and Reproductive Health, Reproductive Epidemiology Research Center, Royan Institute for Reproductive Biomedicine , ACECR, Tehran, Iran
| | - Christiani Andrade Amorim
- 3 Pole de Recherche en Gynecologie, Institut de Recherche Experimentale et Clinique, Universite Catholique de Louvain , Brussels, Belgium
| | - Bita Ebrahimi
- 1 Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine , ACECR, Tehran, Iran
| |
Collapse
|
49
|
Asaduzzman M, Cui X, Zhang H, Young F. Three Dimensional <i>In Vitro</i> Culture of Murine Secondary Follicles in a Defined Synthetic Matrix. ACTA ACUST UNITED AC 2018. [DOI: 10.4236/jbnb.2018.93014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
50
|
Zareifard N, Soleimani A, Talaei-Khozani T, Bahmanpour S. Improved BALB/c mice granulosa cell functions using purified alginate scaffold. IRANIAN JOURNAL OF VETERINARY RESEARCH 2018; 19:182-188. [PMID: 30349564 PMCID: PMC6184026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 04/03/2018] [Accepted: 05/05/2018] [Indexed: 06/08/2023]
Abstract
Alginate, a non-toxic polysaccharide isolated from brown algae, is a widely used 3-dimensional (3D) porous scaffold for the granulosa cell and follicle encapsulation. However, impurities in commercial alginate can lead to alginate biocompatibility reduction. The aim of this study was to evaluate in vitro behavior of the granulosa cells seeded on the purified alginate in varying concentrations compared with matched non-purified ones. We produced a purified alginate using a simple and efficient method. Then, the granulosa cells from mice were isolated and seeded in various concentrations of (0.5%, 1% weight/volume) purified and non-purified alginate. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay was used on the 3rd, 5th and the 8th days of culture as an index of cell viability and proliferation. Furthermore, the secreted estradiol, progesterone and alkaline phosphatase enzyme (ALP) were measured in the granulosa cells culture media using radioimmunoassay kits. The cells cultured on purified and low concentration alginate showed a higher proliferation rate, sex hormone production and ALP activity. The results confirmed the impact of the alginate hydrogel properties on proliferative rate and function of granulosa cells in a 3D culture system.
Collapse
Affiliation(s)
- N. Zareifard
- Stem Cell Research Laboratory, Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - A. Soleimani
- Ph.D. Student in Anatomy, Stem Cell Research Laboratory, Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - T. Talaei-Khozani
- Stem Cell Research Laboratory, Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - S. Bahmanpour
- Ph.D. Student in Anatomy, Stem Cell Research Laboratory, Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|