1
|
Gotschel F, Sonigo C, Becquart C, Sellami I, Mayeur A, Grynberg M. New Insights on In Vitro Maturation of Oocytes for Fertility Preservation. Int J Mol Sci 2024; 25:10605. [PMID: 39408934 PMCID: PMC11477201 DOI: 10.3390/ijms251910605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/20/2024] Open
Abstract
In the last decade, the evolution of oncofertility has sparked a resurgence of interest in in vitro maturation (IVM) due to its suitability in certain oncological scenarios where controlled ovarian hyperstimulation may not be feasible. The retrieval of immature cumulus-oocyte complexes from small antral follicles, regardless of the menstrual cycle phase, presents a swift opportunity to vitrify mature oocytes or embryos post-IVM in urgent situations or when stimulation is not advisable. Harvesting immature cumulus-oocyte complexes and immature oocytes can be achieved transvaginally or directly in the laboratory from extracorporeal ovarian tissue. Although IVM has transitioned from an experimental status due to safety validations, it relies on the intricate process of oocyte maturation. Despite successful live births resulting from IVM in fertility preservation contexts, the comparatively lower developmental competence of in vitro matured oocytes highlights the necessity to enhance IVM culture systems. Recent advancements in IVM systems hold promise in bolstering oocyte competence post-IVM, thereby narrowing the gap between IVM and outcomes from ovarian stimulation. Additionally, for optimizing the chances of conception in cancer survivors, the combination of IVM and ovarian tissue cryopreservation stands as the favored choice when ovarian stimulation is unfeasible.
Collapse
Affiliation(s)
- Flavie Gotschel
- Department of Reproductive Medicine and Fertility Preservation, Université Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Antoine Beclère Hospital, 92140 Clamart, France; (F.G.); (C.B.); (I.S.)
| | - Charlotte Sonigo
- Department of Reproductive Medicine and Fertility Preservation, Université Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Antoine Beclère Hospital, 92140 Clamart, France; (F.G.); (C.B.); (I.S.)
- Inserm, Physiologie et Physiopathologie Endocrinienne, Université Paris-Saclay, 94276 Le Kremlin-Bicêtre, France
| | - Celeste Becquart
- Department of Reproductive Medicine and Fertility Preservation, Université Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Antoine Beclère Hospital, 92140 Clamart, France; (F.G.); (C.B.); (I.S.)
| | - Ines Sellami
- Department of Reproductive Medicine and Fertility Preservation, Université Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Antoine Beclère Hospital, 92140 Clamart, France; (F.G.); (C.B.); (I.S.)
| | - Anne Mayeur
- Histology-Embryology-Cytogenetic Laboratory, Université Paris-Saclay, Assistance Publique Hôpitaux de Paris, Antoine Beclère Hospital, 92140 Clamart, France;
| | - Michael Grynberg
- Department of Reproductive Medicine and Fertility Preservation, Université Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Antoine Beclère Hospital, 92140 Clamart, France; (F.G.); (C.B.); (I.S.)
- Department of Reproductive Medicine and Fertility Preservation, Hôpitaux Universitaires Paris-Seine-Saint-Denis, Assistance Publique-Hôpitaux de Paris, Jean Verdier Hospital, 93143 Bondy, France
- Unité Inserm U1133, Université Paris-Diderot, 75013 Paris, France
| |
Collapse
|
2
|
Ebrahimi M, Dattena M, Mara L, Pasciu V, Sotgiu FD, Chessa F, Luciano AM, Berlinguer F. In vitro production of meiotically competent oocytes from early antral follicles in sheep. Theriogenology 2024; 226:253-262. [PMID: 38950486 DOI: 10.1016/j.theriogenology.2024.06.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/03/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024]
Abstract
The potential of using long in vitro culture (LIVC) of cumulus-oocyte complexes (COCs) from early antral follicles (EAFs) as an assisted reproductive technology in cattle has shown promising results. This study explored the feasibility of applying this technology to sheep as seasonal breeding animals. Ovaries from sheep were collected during both the breeding and non-breeding seasons. COCs were isolated from EAFs (350-450 μm) and cultured in TCM199 medium supplemented with 0.15 μg/mL Zn sulfate, 10-4IU/mL FSH, 10 ng/mL estradiol, 50 ng/mL testosterone, 50 ng/mL progesterone, and 5 μM Cilostamide. After five days of LIVC, the COCs were submitted to an in vitro maturation procedure. The results indicate successful in vitro development of COCs, evidenced by a significant increase in oocyte diameter (p < 0.000) and the preservation of gap junction communication between oocyte and cumulus cells. The gradual uncoupling was accompanied by a progressive chromatin transition from the non-surrounded nucleolus (NSN) to the surrounded nucleolus (SN) (p < 0.000), coupled with a gradual decrease in global transcriptional activity and an increase in oocyte meiotic competence (p < 0.000). Maintenance of oocyte-cumulus investment architecture, viability, and metaphase II capability was significantly higher in COCs collected during the breeding season (p < 0.000), suggesting higher quality than those obtained during the non-breeding season. In conclusion, our study confirms LIVC feasibility in sheep, emphasizing increased effectiveness during the breeding season in isolating higher-quality COCs from EAFs. These findings can influence improving the LIVC system in mammals with seasonal reproduction.
Collapse
Affiliation(s)
- Mohammadreza Ebrahimi
- Department of Veterinary Medicine, University of Sassari, Via Vienna 2, Sassari, Italy; Department of Animal Science, Agricultural Research Agency of Sardinia, 07100, Sassari, Italy.
| | - Maria Dattena
- Department of Animal Science, Agricultural Research Agency of Sardinia, 07100, Sassari, Italy
| | - Laura Mara
- Department of Animal Science, Agricultural Research Agency of Sardinia, 07100, Sassari, Italy.
| | - Valeria Pasciu
- Department of Veterinary Medicine, University of Sassari, Via Vienna 2, Sassari, Italy
| | - Francesca D Sotgiu
- Department of Veterinary Medicine, University of Sassari, Via Vienna 2, Sassari, Italy
| | - Fabrizio Chessa
- Department of Animal Science, Agricultural Research Agency of Sardinia, 07100, Sassari, Italy
| | - Alberto Maria Luciano
- Reproductive and Developmental Biology Laboratory (ReDBioLab), Department of Veterinary Medicine and Animal Sciences, University of Milan, Via Dell'Università, 6 - 26900, Lodi, Italy
| | - Fiammetta Berlinguer
- Department of Veterinary Medicine, University of Sassari, Via Vienna 2, Sassari, Italy
| |
Collapse
|
3
|
Amaral VLL, Nunes JL, Salvador RA, Senn AP, dos Santos TG. In vitro culture of mechanically isolated murine primary follicles in the presence of human platelet lysate PLTMax. JBRA Assist Reprod 2024; 28:410-417. [PMID: 38446749 PMCID: PMC11349267 DOI: 10.5935/1518-0557.20240008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 01/21/2024] [Indexed: 03/08/2024] Open
Abstract
OBJECTIVE To develop a system for the culture of murine preantral ovarian follicles using Human Serum Albumin (HSA) and Human Platelet Lysate (PLTMax). METHODS Mechanically isolated preantral follicles (N=146) were obtained from Swiss mice and cultured in DMEM:F12 medium for ten days in a 96-well plate with conical bottom. The medium was supplemented with penicillin, streptomycin, and equine chorionic gonadotropin. Additional proteins were tested in 4 test groups: G1: human serum albumin (HSA), G2: human platelet lysate (PLTM), and G3 and G4: HSA + PLTMax at lower and higher concentrations, respectively. Cellular vitality and oocyte morphology were evaluated on day 11 of culture. RESULTS The highest follicular growth (3.4 fold) was achieved in HSA (G1), while a significantly lower (1.8 fold) growth was achieved in the presence of PLTM (G2, G4) and even further reduced (1.2 fold) when HSA and PLTM were combined (G3). Cellular vitality was close to 70-80% among the four groups, and the highest number of intact oocytes were found in G1. CONCLUSIONS PLTM did not improve follicular development and oocyte maturation compared to HSA but preserved cell vitality.
Collapse
Affiliation(s)
| | - Jhuly Laurentino Nunes
- Laboratory of reproductive biology, University of Vale do
Itajaí (UNIVALI), Itajaí, SC, Brazil
| | - Rafael Alonso Salvador
- Laboratory of reproductive biology, University of Vale do
Itajaí (UNIVALI), Itajaí, SC, Brazil
| | - Alfred Paul Senn
- Department of Genetic Medicine and Development, University of
Geneva, Geneva, Switzerland
| | | |
Collapse
|
4
|
Blevins GM, Flanagan CL, Kallakuri SS, Meyer OM, Nimmagadda L, Hatch JD, Shea SA, Padmanabhan V, Shikanov A. Quantification of follicles in human ovarian tissue using image processing software and trained artificial intelligence†. Biol Reprod 2024; 110:1086-1099. [PMID: 38537569 PMCID: PMC11180617 DOI: 10.1093/biolre/ioae048] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/12/2024] [Accepted: 03/19/2024] [Indexed: 06/18/2024] Open
Abstract
Cancer survival rates in prepubertal girls and young women have risen in recent decades due to increasingly efficient treatments. However, many such treatments are gonadotoxic, causing premature ovarian insufficiency, loss of fertility, and ovarian endocrine function. Implantation of donor ovarian tissue encapsulated in immune-isolating capsules is a promising method to restore physiological endocrine function without immunosuppression or risk of reintroducing cancer cells harbored by the tissue. The success of this approach is largely determined by follicle density in the implanted ovarian tissue, which is analyzed manually from histologic sections and necessitates specialized, time-consuming labor. To address this limitation, we developed a fully automated method to quantify follicle density that does not require additional coding. We first analyzed ovarian tissue from 12 human donors between 16 and 37 years old using semi-automated image processing with manual follicle annotation and then trained artificial intelligence program based on follicle identification and object classification. One operator manually analyzed 102 whole slide images from serial histologic sections. Of those, 77 images were assessed by a second manual operator, followed with an automated method utilizing artificial intelligence. Of the 1181 follicles the control operator counted, the comparison operator counted 1178, and the artificial intelligence counted 927 follicles with 80% of those being correctly identified as follicles. The three-stage artificial intelligence pipeline finished 33% faster than manual annotation. Collectively, this report supports the use of artificial intelligence and automation to select tissue donors and grafts with the greatest follicle density to ensure graft longevity for premature ovarian insufficiency treatment.
Collapse
Affiliation(s)
- Gabrielle M Blevins
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Colleen L Flanagan
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Sridula S Kallakuri
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Owen M Meyer
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Likitha Nimmagadda
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - James D Hatch
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Sydney A Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Vasantha Padmanabhan
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
| | - Ariella Shikanov
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
- Department of Macromolecular Science and Engineering, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
5
|
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
|
6
|
Li J, Fan H, Liu W, Zhang J, Xiao Y, Peng Y, Yang W, Liu W, He Y, Qin L, Ma X, Li J. Mesenchymal stem cells promote ovarian reconstruction in mice. Stem Cell Res Ther 2024; 15:115. [PMID: 38650029 PMCID: PMC11036642 DOI: 10.1186/s13287-024-03718-z] [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: 09/21/2023] [Accepted: 04/07/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Studies have shown that chemotherapy and radiotherapy can cause premature ovarian failure and loss of fertility in female cancer patients. Ovarian cortex cryopreservation is a good choice to preserve female fertility before cancer treatment. Following the remission of the disease, the thawed ovarian tissue can be transplanted back and restore fertility of the patient. However, there is a risk to reintroduce cancer cells in the body and leads to the recurrence of cancer. Given the low success rate of current in vitro culture techniques for obtaining mature oocytes from primordial follicles, an artificial ovary with primordial follicles may be a good way to solve this problem. METHODS In the study, we established an artificial ovary model based on the participation of mesenchymal stem cells (MSCs) to evaluate the effect of MSCs on follicular development and oocyte maturation. P2.5 mouse ovaries were digested into single cell suspensions and mixed with bone marrow derived mesenchymal stem cells (BM-MSCs) at a 1:1 ratio. The reconstituted ovarian model was then generated by using phytohemagglutinin. The phenotype and mechanism studies were explored by follicle counting, immunohistochemistry, immunofluorescence, in vitro maturation (IVM), in vitro fertilization (IVF), real-time quantitative polymerase chain reaction (RT-PCR), and Terminal-deoxynucleotidyl transferase mediated nick end labeling(TUNEL) assay. RESULTS Our study found that the addition of BM-MSCs to the reconstituted ovary can enhance the survival of oocytes and promote the growth and development of follicles. After transplanting the reconstituted ovaries under kidney capsules of the recipient mice, we observed normal folliculogenesis and oocyte maturation. Interestingly, we found that BM-MSCs did not contribute to the formation of follicles in ovarian aggregation, nor did they undergo proliferation during follicle growth. Instead, the cells were found to be located around growing follicles in the reconstituted ovary. When theca cells were labeled with CYP17a1, we found some overlapped staining with green fluorescent protein(GFP)-labeled BM-MSCs. The results suggest that BM-MSCs may participate in directing the differentiation of theca layer in the reconstituted ovary. CONCLUSIONS The presence of BM-MSCs in the artificial ovary was found to promote the survival of ovarian cells, as well as facilitate follicle formation and development. Since the cells didn't proliferate in the reconstituted ovary, this discovery suggests a potential new and safe method for the application of MSCs in clinical fertility preservation by enhancing the success rate of cryo-thawed ovarian tissues after transplantation.
Collapse
Affiliation(s)
- Jiazhao Li
- State Key Laboratory of Reproductive Medicine and Offspring health, Nanjing Medical University, 210029, Nanjing, China
- Scientific Research Department, Wannan Medical College, 241002, Wuhu, China
| | - Haonan Fan
- State Key Laboratory of Reproductive Medicine and Offspring health, Nanjing Medical University, 210029, Nanjing, China
| | - Wei Liu
- State Key Laboratory of Reproductive Medicine and Offspring health, Nanjing Medical University, 210029, Nanjing, China
| | - Jing Zhang
- State Key Laboratory of Reproductive Medicine and Offspring health, Nanjing Medical University, 210029, Nanjing, China
| | - Yue Xiao
- State Key Laboratory of Reproductive Medicine and Offspring health, Nanjing Medical University, 210029, Nanjing, China
- Center of Reproductive Medicine, The First Affiliated Hospital of Zhejiang University School of Medicine, 310003, Hangzhou, China
| | - Yue Peng
- State Key Laboratory of Reproductive Medicine and Offspring health, Nanjing Medical University, 210029, Nanjing, China
- Pathology Department, Nanjing Kingmed Medical Laboratory Co.,Ltd., 210032, Nanjing, China
| | - Weijie Yang
- State Key Laboratory of Reproductive Medicine and Offspring health, Nanjing Medical University, 210029, Nanjing, China
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Zhejiang University School of Medicine, 310016, Hangzhou, China
| | - Wenwen Liu
- State Key Laboratory of Reproductive Medicine and Offspring health, Nanjing Medical University, 210029, Nanjing, China
- Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), 21003, Nanjing, China
| | - Yuanlin He
- State Key Laboratory of Reproductive Medicine and Offspring health, Nanjing Medical University, 210029, Nanjing, China
| | - Lianju Qin
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center of Clinical Reproductive Medicine, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China.
| | - Xiang Ma
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center of Clinical Reproductive Medicine, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China.
- Prenatal Diagnosis Department, First Affiliated Hospital, Nanjing Medical University, 210029, Nanjing, China.
| | - Jing Li
- State Key Laboratory of Reproductive Medicine and Offspring health, Nanjing Medical University, 210029, Nanjing, China.
| |
Collapse
|
7
|
Jones ASK, Hannum DF, Machlin JH, Tan A, Ma Q, Ulrich ND, Shen YC, Ciarelli M, Padmanabhan V, Marsh EE, Hammoud S, Li JZ, Shikanov A. Cellular atlas of the human ovary using morphologically guided spatial transcriptomics and single-cell sequencing. SCIENCE ADVANCES 2024; 10:eadm7506. [PMID: 38578993 PMCID: PMC10997207 DOI: 10.1126/sciadv.adm7506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 03/04/2024] [Indexed: 04/07/2024]
Abstract
The reproductive and endocrine functions of the ovary involve spatially defined interactions among specialized cell populations. Despite the ovary's importance in fertility and endocrine health, functional attributes of ovarian cells are largely uncharacterized. Here, we profiled >18,000 genes in 257 regions from the ovaries of two premenopausal donors to examine the functional units in the ovary. We also generated single-cell RNA sequencing data for 21,198 cells from three additional donors and identified four major cell types and four immune cell subtypes. Custom selection of sampling areas revealed distinct gene activities for oocytes, theca, and granulosa cells. These data contributed panels of oocyte-, theca-, and granulosa-specific genes, thus expanding the knowledge of molecular programs driving follicle development. Serial samples around oocytes and across the cortex and medulla uncovered previously unappreciated variation of hormone and extracellular matrix remodeling activities. This combined spatial and single-cell atlas serves as a resource for future studies of rare cells and pathological states in the ovary.
Collapse
Affiliation(s)
- Andrea S. K. Jones
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - D. Ford Hannum
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Jordan H. Machlin
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, USA
| | - Ansen Tan
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Qianyi Ma
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
| | - Nicole D. Ulrich
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
| | - Yu-chi Shen
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Maria Ciarelli
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Vasantha Padmanabhan
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Erica E. Marsh
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
| | - Sue Hammoud
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
- Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - Jun Z. Li
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
| | - Ariella Shikanov
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
8
|
Zha H, Yang X, Jiang F, Chen Y, Liang Y, Zhang Z, Yang J. Interleukin-6 Concentration in Single-Embryo Medium Is Associated with Blastocyst Formation. Reprod Sci 2024; 31:1139-1145. [PMID: 38040897 DOI: 10.1007/s43032-023-01414-6] [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: 07/01/2023] [Accepted: 11/16/2023] [Indexed: 12/03/2023]
Abstract
IL-6 plays an important role in oogenesis in humans. However, at the preimplantation stage, IL-6 production and the role in embryo development remain unclear. In this study, IL-6 concentrations in single-embryo media were analyzed. In addition, the association between IL-6 production and blastocyst formation was investigated. Single-embryo culture media from 194 embryos were collected on day 6 after fertilization and divided into four groups according to the developmental stage of the corresponding embryo, as follows: cleavage stage group, morula-early blastocyst group, unavailable full blastocyst group, and available full blastocyst group. IL-6 concentrations were significantly lower in the cleavage stage group than in the morula-early blastocyst group (p = 0.009), in the unavailable full blastocyst group (p = 0.003), and in the available full blastocyst group (p < 0.001). Logistic regression analysis showed that IL-6 concentration in single-embryo medium was significantly associated with blastocyst formation (odds ratios β1 = 1.876, 95% CI 1.433 to 2.644, p < 0.0001). Therefore, IL-6 was produced by human preimplantation embryos throughout the preimplantation stage and may play a role in embryo development.
Collapse
Affiliation(s)
- Hao Zha
- Reproductive Center the Second Affiliated Hospital of Kunming Medical University, Dianmian Road 374, Kunming, 650101, Yunnan, China
| | - Xiaolin Yang
- Reproductive Center the Second Affiliated Hospital of Kunming Medical University, Dianmian Road 374, Kunming, 650101, Yunnan, China
| | - Fangjie Jiang
- Reproductive Center the Second Affiliated Hospital of Kunming Medical University, Dianmian Road 374, Kunming, 650101, Yunnan, China
| | - Yabin Chen
- Reproductive Center the Second Affiliated Hospital of Kunming Medical University, Dianmian Road 374, Kunming, 650101, Yunnan, China
| | - Yu Liang
- Reproductive Center the Second Affiliated Hospital of Kunming Medical University, Dianmian Road 374, Kunming, 650101, Yunnan, China
| | - Zhaoqin Zhang
- Reproductive Center the Second Affiliated Hospital of Kunming Medical University, Dianmian Road 374, Kunming, 650101, Yunnan, China
| | - Jie Yang
- Reproductive Center the Second Affiliated Hospital of Kunming Medical University, Dianmian Road 374, Kunming, 650101, Yunnan, China.
| |
Collapse
|
9
|
Ferronato GDA, Vit FF, da Silveira JC. 3D culture applied to reproduction in females: possibilities and perspectives. Anim Reprod 2024; 21:e20230039. [PMID: 38510565 PMCID: PMC10954237 DOI: 10.1590/1984-3143-ar2023-0039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 12/13/2023] [Indexed: 03/22/2024] Open
Abstract
In vitro cell culture is a well-established technique present in numerous laboratories in diverse areas. In reproduction, gametes, embryos, and reproductive tissues, such as the ovary and endometrium, can be cultured. These cultures are essential for embryo development studies, understanding signaling pathways, developing drugs for reproductive diseases, and in vitro embryo production (IVP). Although many culture systems are successful, they still have limitations to overcome. Three-dimensional (3D) culture systems can be close to physiological conditions, allowing greater interaction between cells and cells with the surrounding environment, maintenance of the cells' natural morphology, and expression of genes and proteins such as in vivo. Additionally, three-dimensional culture systems can stimulated extracellular matrix generating responses due to the mechanical force produced. Different techniques can be used to perform 3D culture systems, such as hydrogel matrix, hanging drop, low attachment surface, scaffold, levitation, liquid marble, and 3D printing. These systems demonstrate satisfactory results in follicle culture, allowing the culture from the pre-antral to antral phase, maintaining the follicular morphology, and increasing the development rates of embryos. Here, we review some of the different techniques of 3D culture systems and their applications to the culture of follicles and embryos, bringing new possibilities to the future of assisted reproduction.
Collapse
Affiliation(s)
| | - Franciele Flores Vit
- Faculdade de Zootecnia e Engenharia de Alimentos, Universidade de São Paulo, Pirassununga, SP, Brasil
| | | |
Collapse
|
10
|
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
|
11
|
Khaleghi S, Eivazkhani F, Tavana S, Moini A, Novin MG, Stoyan P, Nazarian H, Fathi R. Follicular reconstruction and neo-oogenesis in xenotransplantation of human ovarian isolated cells derived from chemotherapy-induced POF patients. J Biol Eng 2023; 17:70. [PMID: 37986177 PMCID: PMC10662631 DOI: 10.1186/s13036-023-00384-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 10/17/2023] [Indexed: 11/22/2023] Open
Abstract
BACKGROUND Developing new strategies to restore fertility in patients with chemotherapy-induced Premature Ovarian Failure (Chemo-POF) is important. We aimed to construct an Artificial Ovary (AO) by seeding Human Ovarian Cortical Cells (HOCCs) into Human ovarian Decellularized Cortical Tissue (DCT). We assessed the AO's ability to produce new ovarian follicles following xenotransplantation to NMRI mice. MATERIAL AND METHODS The DCTs were prepared, and cell removal was confirmed through DNA content, MTT assay, DAPI and H&E staining. Next, HOCCs were isolated from both Chemo-POF and Trans (as a control group) ovarian patients. The HOCCs were characterized using immunostaining (FRAGILIS, Vimentin, and Inhibin α) and real time PCR (DDX4, STELLA, FRAGILIS, Vimentin, FSH-R, KI67) assays. The HOCCs were then seeded into the DCTs and cultured for one week to construct an AO, which was subsequently xenotransplanted into the mice. The existence of active follicles within the AO was studied with H&E and immunofluorescence (GDF9) staining, Real-time PCR (GDF9, ZP3) and hormone analysis (Estradiol, FSH and AMH). RESULTS The results of gene expression and immunostaining showed that 85-86% of the isolated cells from both Trans and Chemo-POF groups were positive for vimentin, while 2-5% were granulosa cells and OSCs were less than 3%. After xenotransplantation, histological study confirmed the presence of morphologically healthy reconstructed human ovarian follicles. Additionally, immunofluorescence staining of GDF9 and hormonal assay confirmed the presence of secretory-active follicles on the AO. CONCLUSION Our findings demonstrate that an artificial ovary produced by seeding HOCCs on DCT can support HOCCs proliferation as well as neo-oogenesis, and enable sex hormone secretion following xenotransplantation.
Collapse
Affiliation(s)
- Sara Khaleghi
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farideh Eivazkhani
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Somayeh Tavana
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Ashraf Moini
- Department of Endocrinology and Female Infertility, Royan Institute of Reproductive Biomedicine, ACECR, Tehran, Iran
- Breast Disease Research Center (BDRC), Tehran University of Medical Science, Tehran, Iran
| | - Marefat Ghaffari Novin
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Petkov Stoyan
- Platform Degenerative Diseases, German Primate Center, GmbH, Leibniz Institute for Primate Research, Göttingen, 37077, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, 37077, Germany
| | - Hamid Nazarian
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Rouhollah Fathi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.
| |
Collapse
|
12
|
Ma Y, Jiang XD, Zhang DW, Zi XD. Molecular characterization and effects of the TGIF1 gene on proliferation and steroidogenesis in yak (Bos grunniens) granulosa cells. Theriogenology 2023; 211:224-231. [PMID: 37660474 DOI: 10.1016/j.theriogenology.2023.08.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/26/2023] [Accepted: 08/28/2023] [Indexed: 09/05/2023]
Abstract
TG interaction factor 1 (TGIF1) plays a major role in transcriptional inhibition and suppression of TGF-β signaling, but its functional roles in granulosa cells (GCs) have not been elucidated; in particular, there is no information about the yak (Bos grunniens) TGIF1 gene. Therefore, the objectives of this study were to clone yak TGIF1 and investigate TGIF1 functions in yak GCs. RT‒PCR results showed that the coding region of yak TGIF1 is 759 bp and encodes 252 amino acids. Its nucleotide sequence showed 85.24-99.74% similarity to mouse, human, pig, goat and cattle homologous genes. To explore the functional roles of TGIF1, we studied proliferation, apoptosis, cell cycle progression, steroidogenesis and the expression levels of related genes in yak GCs transfected with small interfering RNA specific to TGIF1. The results showed that TGIF1 knockdown promoted proliferation and cell cycle progression and inhibited apoptosis and estradiol (E2) and progesterone (P4) production in cultured yak GCs. Conversely, TGIF1 overexpression inhibited proliferation and cell cycle progression and stimulated apoptosis and E2 and P4 production. In addition, these functional changes in yak GCs were observed parallel to the expression changes in genes involved in the cell cycle (PCNA, CDK2, CCND1, CCNE1, CDK4 and P53), apoptosis (BCL2, BAX and CASPASE3), and steroidogenesis (CYP11A1, 3β-HSD and StAR). In conclusion, TGIF1 was relatively conserved in the course of animal evolution. TGIF1 inhibited GC viability and stimulated apoptosis and the secretion of E2 and P4 by yak GCs. Our results will help to reveal the mechanism underlying yak follicular development and improve the reproductive efficiency of female yaks.
Collapse
Affiliation(s)
- Yao Ma
- The Key Laboratory for Animal Science of National Ethnic Affairs Commission, Southwest Minzu University, Chengdu, 610041, PR China
| | - Xu-Dong Jiang
- The Key Laboratory for Animal Science of National Ethnic Affairs Commission, Southwest Minzu University, Chengdu, 610041, PR China
| | - Da-Wei Zhang
- College of Food Science and Technology, Southwest Minzu University, Chengdu, 610041, PR China.
| | - Xiang-Dong Zi
- The Key Laboratory for Animal Science of National Ethnic Affairs Commission, Southwest Minzu University, Chengdu, 610041, PR China.
| |
Collapse
|
13
|
Telfer EE, Grosbois J, Odey YL, Rosario R, Anderson RA. Making a good egg: human oocyte health, aging, and in vitro development. Physiol Rev 2023; 103:2623-2677. [PMID: 37171807 PMCID: PMC10625843 DOI: 10.1152/physrev.00032.2022] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 05/03/2023] [Accepted: 05/06/2023] [Indexed: 05/13/2023] Open
Abstract
Mammalian eggs (oocytes) are formed during fetal life and establish associations with somatic cells to form primordial follicles that create a store of germ cells (the primordial pool). The size of this pool is influenced by key events during the formation of germ cells and by factors that influence the subsequent activation of follicle growth. These regulatory pathways must ensure that the reserve of oocytes within primordial follicles in humans lasts for up to 50 years, yet only approximately 0.1% will ever be ovulated with the rest undergoing degeneration. This review outlines the mechanisms and regulatory pathways that govern the processes of oocyte and follicle formation and later growth, within the ovarian stroma, through to ovulation with particular reference to human oocytes/follicles. In addition, the effects of aging on female reproductive capacity through changes in oocyte number and quality are emphasized, with both the cellular mechanisms and clinical implications discussed. Finally, the details of current developments in culture systems that support all stages of follicle growth to generate mature oocytes in vitro and emerging prospects for making new oocytes from stem cells are outlined.
Collapse
Affiliation(s)
- Evelyn E Telfer
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Johanne Grosbois
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Yvonne L Odey
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Roseanne Rosario
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Richard A Anderson
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
14
|
Li C, Zhou M, He X, Di R, Zhang Z, Ren C, Liu Q, Chu M. Comparative proteomics of ovaries elucidated the potential targets related to ovine prolificacy. Front Vet Sci 2023; 10:1096762. [PMID: 37675075 PMCID: PMC10477366 DOI: 10.3389/fvets.2023.1096762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 08/02/2023] [Indexed: 09/08/2023] Open
Abstract
Small Tail Han (STH) sheep, a unique Chinese breed, is recognized for its early maturity, year-round estrus, and prolificacy. However, the molecular mechanism of its high prolificacy has not been fully elucidated. The Proteomics approach is feasible and effective to reveal the proteins involved in the complex physiological processes of any organism. Given this, we performed the protein expression profiling of ovarian tissues during the luteal phase using polytocous STH sheep (litter size ≥2, three consecutive lambings) and monotocous STH sheep (litter size =1, three consecutive lambings) (PL vs. ML), and the follicular phase using polytocous STH sheep (litter size ≥2, three consecutive lambings) and monotocous STH sheep (litter size =1, three consecutive lambings) (PF vs. MF), respectively. Parallel Reaction Monitoring (PRM) was conducted to validate the differentially abundant proteins (DAPs). The tandem mass tag (TMT) quantitative proteomic results showed that a total of 5,237 proteins were identified, of which 49 and 44 showed differential abundance in the PL vs. ML and PF vs. MF groups, respectively. Enrichments analyses indicated that the DAPs including TIA1 cytotoxic granule-associated RNA-binding protein-like 1 (TIAL1), nicotinamide phosphoribosyltransferase (NAMPT), and cellular retinoic acid-binding protein 1 (CRABP1) were enriched at the luteal phase, while TIAL1, inhibin beta-a-subunit (A2ICA4), and W5PG55 were enriched at the follicular phase, potentially mediating reproductive processes in polytocous ewes. Furthermore, six DAPs were verified using PRM, confirming the accuracy of the TMT data acquired in this study. Together, our work expanded the database of indigenous sheep breeds and provided new ovarian candidate molecular targets, which will help in the study of the genetic mechanisms of ovine prolificacy.
Collapse
Affiliation(s)
- Chunyan Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
- Yunnan Provincial Engineering Laboratory of Animal Genetic Resource Conservation and Germplasm Enhancement, Yunnan Academy of Animal Husbandry and Veterinary Sciences, Kunming, China
| | - Mei Zhou
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
- Key Laboratory of Pig Molecular Quantitative Genetics of Anhui Academy of Agricultural Sciences, Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei, China
| | - Xiaoyun He
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Ran Di
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zijun Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Chunhuan Ren
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Qiuyue Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Mingxing Chu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
15
|
Wang X, Zhou S, Wu Z, Liu R, Ran Z, Liao J, Shi H, Wang F, Chen J, Liu G, Liang A, Yang L, Zhang S, Li X, He C. The FSH-mTOR-CNP signaling axis initiates follicular antrum formation by regulating tight junction, ion pumps, and aquaporins. J Biol Chem 2023; 299:105015. [PMID: 37414146 PMCID: PMC10424218 DOI: 10.1016/j.jbc.2023.105015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/20/2023] [Accepted: 06/28/2023] [Indexed: 07/08/2023] Open
Abstract
The initial formation of the follicular antrum (iFFA) serves as a dividing line between gonadotropin-independent and gonadotropin-dependent folliculogenesis, enabling the follicle to sensitively respond to gonadotropins for its further development. However, the mechanism underlying iFFA remains elusive. Herein, we reported that iFFA is characterized by enhanced fluid absorption, energy consumption, secretion, and proliferation and shares a regulatory mechanism with blastula cavity formation. By use of bioinformatics analysis, follicular culture, RNA interference, and other techniques, we further demonstrated that the tight junction, ion pumps, and aquaporins are essential for follicular fluid accumulation during iFFA, as a deficiency of any one of these negatively impacts fluid accumulation and antrum formation. The intraovarian mammalian target of rapamycin-C-type natriuretic peptide pathway, activated by follicle-stimulating hormone, initiated iFFA by activating tight junction, ion pumps, and aquaporins. Building on this, we promoted iFFA by transiently activating mammalian target of rapamycin in cultured follicles and significantly increased oocyte yield. These findings represent a significant advancement in iFFA research, further enhancing our understanding of folliculogenesis in mammals.
Collapse
Affiliation(s)
- Xiaodong Wang
- National Center for International Research on Animal Genetics, Breeding and Reproduction/Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology/Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Shanshan Zhou
- National Center for International Research on Animal Genetics, Breeding and Reproduction/Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology/Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zian Wu
- National Center for International Research on Animal Genetics, Breeding and Reproduction/Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology/Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ruiyan Liu
- National Center for International Research on Animal Genetics, Breeding and Reproduction/Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology/Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zaohong Ran
- National Center for International Research on Animal Genetics, Breeding and Reproduction/Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology/Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Jianning Liao
- National Center for International Research on Animal Genetics, Breeding and Reproduction/Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology/Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Hongru Shi
- National Center for International Research on Animal Genetics, Breeding and Reproduction/Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology/Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Feng Wang
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, USA
| | - Jianguo Chen
- National Center for International Research on Animal Genetics, Breeding and Reproduction/Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology/Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Guoshi Liu
- Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Aixin Liang
- National Center for International Research on Animal Genetics, Breeding and Reproduction/Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology/Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Liguo Yang
- National Center for International Research on Animal Genetics, Breeding and Reproduction/Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology/Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Shujun Zhang
- National Center for International Research on Animal Genetics, Breeding and Reproduction/Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology/Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiang Li
- National Center for International Research on Animal Genetics, Breeding and Reproduction/Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology/Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Changjiu He
- National Center for International Research on Animal Genetics, Breeding and Reproduction/Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Sciences and Technology/Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.
| |
Collapse
|
16
|
Dalman A, Adib S, Amorim CA, Pirjani R, Totonchi M, Valojerdi MR. Co-culture of human cryopreserved fragmented ovarian tissue with theca progenitor cells derived from theca stem cells. J Assist Reprod Genet 2023; 40:1611-1622. [PMID: 37079226 PMCID: PMC10352475 DOI: 10.1007/s10815-023-02799-8] [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: 11/29/2022] [Accepted: 04/05/2023] [Indexed: 04/21/2023] Open
Abstract
PURPOSE Despite the significant advances in the in vitro development of human primordial follicles, it is still a challenging approach with great potential for improvements. Therefore, the present study aimed to investigate the effect of a feeder layer of human theca progenitor cells (hTPCs) on the development of primordial follicles embedded in human ovarian tissue. METHODS Fragments of frozen-thawed ovarian tissue were activated using the vanadate-derivative dipotassium bisperoxo (5-hydroxy-pyridine-2-carboxylic) oxovanadate (V) and kit ligand for 24 h. Then, the specimens were divided into the co-culture and mono-culture groups and were cultured with and without a hTPC feeder layer for 6 days, respectively. Afterward, the follicles were counted and classified, and the hormone levels and expression levels of apoptosis- and folliculogenesis-related genes were assessed. RESULTS Both culture groups showed significant follicle growth (P < 0.05). However, the co-culture group had a significantly higher number of growing follicles compared to the other group (P < 0.05). Moreover, the expression levels of ZP1, ZP2, ZP3, BMP-7, AMH, and GDF9 were significantly higher in the co-culture group compared to the other group (P < 0.05), while the expression levels of P53 and CASP3 were significantly lower (P < 0.05). Also, the concentrations of estradiol, progesterone, testosterone, and androstenedione were significantly higher in the co-culture group compared to the other group (P < 0.05). CONCLUSION The present study results provided novel evidence on the direct role of hTPCs in the growth and development of human primordial follicles. However, there is a need for future studies to illustrate the underlying mechanisms. Schematic summary of the results. According to our results, the expression of ZP1, ZP2, ZP3, and GDF9 in the oocytes, AMH in the granulosa cells, and BMP4 in the theca cells of the co-culture group were significantly higher than those of the mono-culture and non-culture groups, while the expression of apoptotic genes (BAX, CASP3, and P53) was significantly lower. Moreover, the co-culture group showed significantly increased levels of estradiol, progesterone, testosterone, and androstenedione in its culture media compared to the mono-culture groups.
Collapse
Affiliation(s)
- Azam Dalman
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Banihashem Avenue, Resalat Highway, PO Box 19395- 4644, Tehran, Iran.
| | - Samane Adib
- Faculty of Medicine, Department of Anatomical Sciences & Cognitive Neuroscience, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Christiani A Amorim
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale Et Clinique, Université Catholique de Louvain, Avenue Hippocrate 55, Bte. B1.55.03, 1200, Bruxelles, Belgique
| | - Reihaneh Pirjani
- Department of Obstetrics and Gynecology, Arash Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Totonchi
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Mojtaba Rezazadeh Valojerdi
- Department of Anatomy, Faculty of Medical Sciences, Tarbiat Modares University, Jalal-Ale-Ahmad Street, P.O.Box:14115-111, Tehran, Iran.
| |
Collapse
|
17
|
Zhu M, Xu M, Zhang J, Zheng C. The role of Hippo pathway in ovarian development. Front Physiol 2023; 14:1198873. [PMID: 37334049 PMCID: PMC10275494 DOI: 10.3389/fphys.2023.1198873] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 05/22/2023] [Indexed: 06/20/2023] Open
Abstract
The follicle is the functional unit of the ovary, whereby ovarian development is largely dependent on the development of the follicles themselves. The activation, growth, and progression of follicles are modulated by a diverse range of factors, including reproductive endocrine system and multiple signaling pathways. The Hippo pathway exhibits a high degree of evolutionary conservation between both Drosophila and mammalian systems, and is recognized for its pivotal role in regulating cellular proliferation, control of organ size, and embryonic development. During the process of follicle development, the components of the Hippo pathway show temporal and spatial variations. Recent clinical studies have shown that ovarian fragmentation can activate follicles. The mechanism is that the mechanical signal of cutting triggers actin polymerization. This process leads to the disruption of the Hippo pathway and subsequently induces the upregulation of downstream CCN and apoptosis inhibitors, thereby promoting follicle development. Thus, the Hippo pathway plays a crucial role in both the activation and development of follicles. In this article, we focused on the development and atresia of follicles and the function of Hippo pathway in these processes. Additionally, the physiological effects of Hippo pathway in follicle activation are also explored.
Collapse
|
18
|
Han E, Seifer DB. Oocyte Cryopreservation for Medical and Planned Indications: A Practical Guide and Overview. J Clin Med 2023; 12:jcm12103542. [PMID: 37240648 DOI: 10.3390/jcm12103542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Oocyte cryopreservation (OC) is the process in which ovarian follicles are stimulated, the follicular fluid is retrieved, and mature oocytes are isolated and vitrified. Since the first successful pregnancy utilizing previously cryopreserved oocytes in 1986, OC has become increasingly utilized as an option for future biologic children in patients facing gonadotoxic therapies, such as for the treatment of cancer. Planned OC, also termed elective OC, is growing in popularity as a means to circumvent age-related fertility decline. In this narrative review, we describe both medically indicated and planned OC, focusing on the physiology of ovarian follicular loss, OC technique and risks, timing of when OC should be performed, associated financial considerations, and outcomes.
Collapse
Affiliation(s)
- Eric Han
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510, USA
| | - David B Seifer
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
19
|
Hu B, Wang R, Wu D, Long R, Ruan J, Jin L, Ma D, Sun C, Liao S. Prospects for fertility preservation: the ovarian organ function reconstruction techniques for oogenesis, growth and maturation in vitro. Front Physiol 2023; 14:1177443. [PMID: 37250136 PMCID: PMC10213246 DOI: 10.3389/fphys.2023.1177443] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/04/2023] [Indexed: 05/31/2023] Open
Abstract
Today, fertility preservation is receiving more attention than ever. Cryopreservation, which preserves ovarian tissue to preserve fertility in young women and reduce the risk of infertility, is currently the most widely practiced. Transplantation, however, is less feasible for women with blood-borne leukemia or cancers with a high risk of ovarian metastasis because of the risk of cancer recurrence. In addition to cryopreservation and re-implantation of embryos, in vitro ovarian organ reconstruction techniques have been considered as an alternative strategy for fertility preservation. In vitro culture of oocytes in vitro Culture, female germ cells induction from pluripotent stem cells (PSC) in vitro, artificial ovary construction, and ovaria-related organoids construction have provided new solutions for fertility preservation, which will therefore maximize the potential for all patients undergoing fertility preservation. In this review, we discussed and thought about the latest ovarian organ function reconstruction techniques in vitro to provide new ideas for future ovarian disease research and fertility preservation of patients with cancer and premature ovarian failure.
Collapse
Affiliation(s)
- Bai Hu
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Renjie Wang
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Di Wu
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Long
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinghan Ruan
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Jin
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ding Ma
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chaoyang Sun
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shujie Liao
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
20
|
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
|
21
|
Koike H, Harada M, Kusamoto A, Xu Z, Tanaka T, Sakaguchi N, Kunitomi C, Azhary JMK, Takahashi N, Urata Y, Osuga Y. Roles of endoplasmic reticulum stress in the pathophysiology of polycystic ovary syndrome. Front Endocrinol (Lausanne) 2023; 14:1124405. [PMID: 36875481 PMCID: PMC9975510 DOI: 10.3389/fendo.2023.1124405] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/02/2023] [Indexed: 02/17/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine disorder among reproductive-age women, affecting up to 15% of women in this group, and the most common cause of anovulatory infertility. Although its etiology remains unclear, recent research has revealed the critical role of endoplasmic reticulum (ER) stress in the pathophysiology of PCOS. ER stress is defined as a condition in which unfolded or misfolded proteins accumulate in the ER because of an imbalance in the demand for protein folding and the protein-folding capacity of the ER. ER stress results in the activation of several signal transduction cascades, collectively termed the unfolded protein response (UPR), which regulates various cellular activities. In principle, the UPR restores homeostasis and keeps the cell alive. However, if the ER stress cannot be resolved, it induces programmed cell death. ER stress has recently been recognized to play diverse roles in both physiological and pathological conditions of the ovary. In this review, we summarize current knowledge of the roles of ER stress in the pathogenesis of PCOS. ER stress pathways are activated in the ovaries of both a mouse model of PCOS and in humans, and local hyperandrogenism in the follicular microenvironment associated with PCOS is responsible for activating these. The activation of ER stress contributes to the pathophysiology of PCOS through multiple effects in granulosa cells. Finally, we discuss the potential for ER stress to serve as a novel therapeutic target for PCOS.
Collapse
Affiliation(s)
- Hiroshi Koike
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Miyuki Harada
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
- *Correspondence: Miyuki Harada,
| | - Akari Kusamoto
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Zixin Xu
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tsurugi Tanaka
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Nanoka Sakaguchi
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Chisato Kunitomi
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Jerilee M. K. Azhary
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Nozomi Takahashi
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoko Urata
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
22
|
Buckenmeyer MJ, Sukhwani M, Iftikhar A, Nolfi AL, Xian Z, Dadi S, Case ZW, Steimer SR, D’Amore A, Orwig KE, Brown BN. A bioengineered in situ ovary (ISO) supports follicle engraftment and live-births post-chemotherapy. J Tissue Eng 2023; 14:20417314231197282. [PMID: 38029018 PMCID: PMC10656812 DOI: 10.1177/20417314231197282] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 08/10/2023] [Indexed: 12/01/2023] Open
Abstract
Female cancer patients who have undergone chemotherapy have an elevated risk of developing ovarian dysfunction and failure. Experimental approaches to treat iatrogenic infertility are evolving rapidly; however, challenges and risks remain that hinder clinical translation. Biomaterials have improved in vitro follicle maturation and in vivo transplantation in mice, but there has only been marginal success for early-stage human follicles. Here, we developed methods to obtain an ovarian-specific extracellular matrix hydrogel to facilitate follicle delivery and establish an in situ ovary (ISO), which offers a permissive environment to enhance follicle survival. We demonstrate sustainable follicle engraftment, natural pregnancy, and the birth of healthy pups after intraovarian microinjection of isolated exogenous follicles into chemotherapy-treated (CTx) mice. Our results confirm that hydrogel-based follicle microinjection could offer a minimally invasive delivery platform to enhance follicle integration for patients post-chemotherapy.
Collapse
Affiliation(s)
- Michael J Buckenmeyer
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Meena Sukhwani
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Aimon Iftikhar
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alexis L Nolfi
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ziyu Xian
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Srujan Dadi
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zachary W Case
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sarah R Steimer
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Antonio D’Amore
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Fondazione RiMED, Palermo, Italy
| | - Kyle E Orwig
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Bryan N Brown
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
23
|
Distribution of tetraspanins in bovine ovarian tissue and fresh/vitrified oocytes. Histochem Cell Biol 2023; 159:163-183. [PMID: 36242635 PMCID: PMC9922244 DOI: 10.1007/s00418-022-02155-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2022] [Indexed: 11/04/2022]
Abstract
Tetraspanin proteins are mostly known as organizers of molecular complexes on cell membranes, widely expressed on the surface of most nucleated cells. Although tetraspanins participate in many physiological processes of mammals, including reproduction, their relevance to the processes of folliculogenesis and oogenesis has not yet been fully elucidated. We bring new information regarding the distribution of tetraspanins CD9, CD81, CD151, CD82, and CD63 at different stages of follicular development in cattle. The found distribution of tetraspanin CD9, CD63, and integrin alpha V in similar areas of ovarian tissue outlined their possible cooperation. We also describe yet-unknown distribution patterns of CD151, CD82, and CD63 on immature and mature bovine oocytes. The unique localization of tetraspanins CD63 and CD82 in the zona pellucida of bovine oocytes suggested their involvement in transzonal projections. Furthermore, we present an unchanged distribution pattern of the studied tetraspanins in vitrified mature bovine oocytes. The immunofluorescent analysis was supplemented by in silico data addressing tetraspanins expression in the ovarian cells and oocytes across several species. The obtained results suggest that in the study of the oocyte development and potentially the fertilization process of cattle, the role of tetraspanins and integrins should also be taken into account.
Collapse
|
24
|
Biswas A, Ng BH, Prabhakaran VS, Chan CJ. Squeezing the eggs to grow: The mechanobiology of mammalian folliculogenesis. Front Cell Dev Biol 2022; 10:1038107. [PMID: 36531957 PMCID: PMC9756970 DOI: 10.3389/fcell.2022.1038107] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/16/2022] [Indexed: 08/25/2023] Open
Abstract
The formation of functional eggs (oocyte) in ovarian follicles is arguably one of the most important events in early mammalian development since the oocytes provide the bulk genetic and cytoplasmic materials for successful reproduction. While past studies have identified many genes that are critical to normal ovarian development and function, recent studies have highlighted the role of mechanical force in shaping folliculogenesis. In this review, we discuss the underlying mechanobiological principles and the force-generating cellular structures and extracellular matrix that control the various stages of follicle development. We also highlight emerging techniques that allow for the quantification of mechanical interactions and follicular dynamics during development, and propose new directions for future studies in the field. We hope this review will provide a timely and useful framework for future understanding of mechano-signalling pathways in reproductive biology and diseases.
Collapse
Affiliation(s)
- Arikta Biswas
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Boon Heng Ng
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | | | - Chii Jou Chan
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| |
Collapse
|
25
|
Development and Survival of Human Ovarian Cells in Chitosan Hydrogel Micro-Bioreactor. Medicina (B Aires) 2022; 58:medicina58111565. [DOI: 10.3390/medicina58111565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/20/2022] [Accepted: 10/22/2022] [Indexed: 11/06/2022] Open
Abstract
Background and Objectives: To test the long-term ability of human ovarian cortex cells to develop in unconventional culture conditions. Materials and Methods. Ovarian cortex cells from fetuses aged 23 to 39 weeks gestation were cultured for 90 days in hollow chitosan hydrogel micro-bioreactors and concurrently in traditional wells. Various cell-type counts were considered. Results: With intact follicles as a denominator, the percentage of growing intact follicles at Day 0 varied widely between ovaries (0 to 31.7%). This percentage tended to increase or stay relatively constant in bioreactor as in control cultures; it tended more toward an increase over time in bioreactor vs. control cultures. Modeled percentages showed differences (though not significant) in favor of bioreactor cultures (16.12% difference at D50 but only 0.12% difference at D90). With all follicles present as a denominator, the percentage of growing primary and secondary follicles at D0 varied widely between ovaries (0 to 29.3%). This percentage tended to increase over time in bioreactor cultures but to decrease in control cultures. Modeled percentages showed significant differences in favor of bioreactor cultures (8.9% difference at D50 and 11.1% difference at D90). At D50 and D90, there were only few and sparse apoptotic cells in bioreactor cultures vs. no apoptotic cells in control cultures. Conclusions: Over three months, bioreactor folliculogenesis outperformed slightly traditional culture. This is an interesting perspective for follicle preservation and long-term toxicological studies.
Collapse
|
26
|
Di Berardino C, Peserico A, Capacchietti G, Zappacosta A, Bernabò N, Russo V, Mauro A, El Khatib M, Gonnella F, Konstantinidou F, Stuppia L, Gatta V, Barboni B. High-Fat Diet and Female Fertility across Lifespan: A Comparative Lesson from Mammal Models. Nutrients 2022; 14:nu14204341. [PMID: 36297035 PMCID: PMC9610022 DOI: 10.3390/nu14204341] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/10/2022] [Accepted: 10/14/2022] [Indexed: 11/18/2022] Open
Abstract
Female reproduction focuses mainly on achieving fully grown follicles and competent oocytes to be successfully fertilized, as well as on nourishing the developing offspring once pregnancy occurs. Current evidence demonstrates that obesity and/or high-fat diet regimes can perturbate these processes, leading to female infertility and transgenerational disorders. Since the mechanisms and reproductive processes involved are not yet fully clarified, the present review is designed as a systematic and comparative survey of the available literature. The available data demonstrate the adverse influences of obesity on diverse reproductive processes, such as folliculogenesis, oogenesis, and embryo development/implant. The negative reproductive impact may be attributed to a direct action on reproductive somatic and germinal compartments and/or to an indirect influence mediated by the endocrine, metabolic, and immune axis control systems. Overall, the present review highlights the fragmentation of the current information limiting the comprehension of the reproductive impact of a high-fat diet. Based on the incidence and prevalence of obesity in the Western countries, this topic becomes a research challenge to increase self-awareness of dietary reproductive risk to propose solid and rigorous preventive dietary regimes, as well as to develop targeted pharmacological interventions.
Collapse
Affiliation(s)
- Chiara Di Berardino
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Alessia Peserico
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
- Correspondence:
| | - Giulia Capacchietti
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Alex Zappacosta
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Nicola Bernabò
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council, A. Buzzati-Traverso Campus, via E. Ramarini 32, Monterotondo Scalo, 00015 Rome, Italy
| | - Valentina Russo
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Annunziata Mauro
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Mohammad El Khatib
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| | - Francesca Gonnella
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Fani Konstantinidou
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Liborio Stuppia
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Valentina Gatta
- Department of Psychological Health and Territorial Sciences, School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Unit of Molecular Genetics, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Barbara Barboni
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
| |
Collapse
|
27
|
Passos JRS, Guerreiro DD, Otávio KS, Dos Santos-Neto PC, Souza-Neves M, Cuadro F, Nuñez-Olivera R, Crispo M, Vasconcelos FR, Bezerra MJB, Silva RF, Lima LF, Figueiredo JR, Bustamante-Filho IC, Menchaca A, Moura AA. How in vitro maturation changes the proteome of ovine cumulus-oocyte complexes? Mol Reprod Dev 2022; 89:459-470. [PMID: 35901249 DOI: 10.1002/mrd.23638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 05/29/2022] [Accepted: 07/21/2022] [Indexed: 11/11/2022]
Abstract
The present study evaluated the effects of in vitro maturation (IVM) on the proteome of cumulus-oocyte complexes (COCs) from ewes. Extracted COC proteins were analyzed by LC-MS/MS. Differences in protein abundances (p < 0.05) and functional enrichments in immature versus in vitro-matured COCs were evaluated using bioinformatics tools. There were 2550 proteins identified in the COCs, with 89 and 87 proteins exclusive to immature and mature COCs, respectively. IVM caused downregulation of 84 and upregulation of 34 proteins. Major upregulated proteins in mature COCs were dopey_N domain-containing protein, structural maintenance of chromosomes protein, ubiquitin-like modifier-activating enzyme 2. Main downregulated proteins in mature COCs were immunoglobulin heavy constant mu, inter-alpha-trypsin inhibitor heavy chain 2, alpha-2-macroglobulin. Proteins exclusive to mature COCs and upregulated after IVM related to immune response, complement cascade, vesicle-mediated transport, cell cycle, and extracellular matrix organization. Proteins of immature COCs and downregulated after IVM were linked to metabolic processes, immune response, and complement cascade. KEGG pathways and miRNA-regulated genes attributed to downregulated and mature COC proteins related to complement and coagulation cascades, metabolism, humoral response, and B cell-mediated immunity. Thus, IVM influenced the ovine COC proteome. This knowledge supports the future development of efficient IVM protocols for Ovis aries.
Collapse
Affiliation(s)
- José Renato S Passos
- Laboratory of Animal Physiology, Department of Animal Science, Federal University of Ceará, Fortaleza, Brazil
| | - Denise D Guerreiro
- Laboratory of Animal Physiology, Department of Animal Science, Federal University of Ceará, Fortaleza, Brazil
| | - Kamila S Otávio
- Laboratory of Animal Physiology, Department of Animal Science, Federal University of Ceará, Fortaleza, Brazil
| | | | - Marcela Souza-Neves
- Instituto de Reproducción Animal Uruguay, Fundación IRAUy, Montevideo, Uruguay
| | - Federico Cuadro
- Instituto de Reproducción Animal Uruguay, Fundación IRAUy, Montevideo, Uruguay
| | | | - Martina Crispo
- Unidad de Biotecnología en Animales de Laboratorio, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Fábio R Vasconcelos
- Laboratory of Animal Physiology, Department of Animal Science, Federal University of Ceará, Fortaleza, Brazil
| | - Maria Julia B Bezerra
- Laboratory of Animal Physiology, Department of Animal Science, Federal University of Ceará, Fortaleza, Brazil
| | - Renato F Silva
- Laboratory of Manipulation of Oocyte and Preantral Follicles (LAMOFOPA), Ceará State University, Fortaleza, Brazil
| | - Laritza F Lima
- Laboratory of Manipulation of Oocyte and Preantral Follicles (LAMOFOPA), Ceará State University, Fortaleza, Brazil
| | - José Ricardo Figueiredo
- Laboratory of Manipulation of Oocyte and Preantral Follicles (LAMOFOPA), Ceará State University, Fortaleza, Brazil
| | | | - Alejo Menchaca
- Instituto de Reproducción Animal Uruguay, Fundación IRAUy, Montevideo, Uruguay.,Plataforma de Investigación en Salud Animal, Instituto Nacional de Investigación Agropecuaria, Montevideo, Uruguay
| | - Arlindo A Moura
- Laboratory of Animal Physiology, Department of Animal Science, Federal University of Ceará, Fortaleza, Brazil
| |
Collapse
|
28
|
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: 14] [Impact Index Per Article: 7.0] [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
|
29
|
Zi XD, Xiong Y, Wu JB, Gige MT, Zhao SB, Yu ZH, Lu Y, Qiao YS. Dynamic changes in gene expression during follicle development and the efficacy of four timed AI protocols in non-suckling female yaks ( Bos grunniens). Anim Biotechnol 2022:1-7. [PMID: 35133936 DOI: 10.1080/10495398.2022.2030346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
The objectives of the study were to investigate changes in the mRNA expression levels of five genes during antral follicle development and to assess the efficacy of four timed-artificial insemination (TAI) protocols in female yaks (Bos grunniens). RT-qPCR analysis revealed that expression levels were greater for follicle-stimulating hormone receptor and bone morphogenic protein 15 in the small follicle, luteinizing hormone receptor, and kit ligand in the large follicle, and growth differentiation factor 9 in the medium follicle (p < 0.05). Non-suckling yaks were treated as a 7-d CIDR, and PGF2α + eCG at CIDR withdrawal and TAI with frozen yak semen at 56-58 h after PGF2α (PPe-7d); either a 7-d CIDR (PPG-7d) or a 5-d CIDR (PPG-5d), and PGF2α at CIDR withdrawal and TAI + GnRH at 70-72 h after PGF2α; and GnRH treatment on Day 0, followed by PGF2α on Day 7 and TAI + GnRH on Day 9 (GPG-7d). The results showed that the pregnancy rate (P/AI) was greater in PPG-5d than in GPG-7d (p < 0.05), but the P/AI was not different among the other TAI protocols. In conclusion, the expression levels of these genes in follicles are dynamically changed during antral follicle development in yaks. The PPG-5d protocol achieved a greater P/AI.
Collapse
Affiliation(s)
- Xiang-Dong Zi
- The Key Laboratory of Animal Science of State Ethnic Affairs Commission, Southwest Minzu University, Chengdu, China
| | - Yan Xiong
- The Key Laboratory of Animal Science of State Ethnic Affairs Commission, Southwest Minzu University, Chengdu, China
| | - Jin-Bo Wu
- Institute of Animal Science and Veterinary Medicine of Aba Prefecture, Sichuan, China
| | - Mo-Ti Gige
- The Key Laboratory of Animal Science of State Ethnic Affairs Commission, Southwest Minzu University, Chengdu, China
| | - Shou-Bao Zhao
- Datong Yak Breeding Farm of Qinghai Province, Xining, China
| | - Zhong-Hua Yu
- Institute of Animal Science and Veterinary Medicine of Aba Prefecture, Sichuan, China
| | - Yong Lu
- Institute of Animal Science and Veterinary Medicine of Aba Prefecture, Sichuan, China
| | | |
Collapse
|
30
|
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: 3.0] [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
|
31
|
Wu J, Liu Y, Song Y, Wang L, Ai J, Li K. Aging conundrum: A perspective for ovarian aging. Front Endocrinol (Lausanne) 2022; 13:952471. [PMID: 36060963 PMCID: PMC9437485 DOI: 10.3389/fendo.2022.952471] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
Progressive loss of physiological integrity and accumulation of degenerative changes leading to functional impairment and increased susceptibility to diseases are the main features of aging. The ovary, the key organ that maintains female reproductive and endocrine function, enters aging earlier and faster than other organs and has attracted extensive attention from society. Ovarian aging is mainly characterized by the progressive decline in the number and quality of oocytes, the regulatory mechanisms of which have yet to be systematically elucidated. This review discusses the hallmarks of aging to further highlight the main characteristics of ovarian aging and attempt to explore its clinical symptoms and underlying mechanisms. Finally, the intervention strategies related to aging are elaborated, especially the potential role of stem cells and cryopreservation of embryos, oocytes, or ovarian tissue in the delay of ovarian aging.
Collapse
Affiliation(s)
| | | | | | - Lingjuan Wang
- *Correspondence: Kezhen Li, ; Jihui Ai, ; Lingjuan Wang,
| | - Jihui Ai
- *Correspondence: Kezhen Li, ; Jihui Ai, ; Lingjuan Wang,
| | - Kezhen Li
- *Correspondence: Kezhen Li, ; Jihui Ai, ; Lingjuan Wang,
| |
Collapse
|
32
|
Xu J, Zelinski MB. Oocyte quality following in vitro follicle development†. Biol Reprod 2021; 106:291-315. [PMID: 34962509 PMCID: PMC9004734 DOI: 10.1093/biolre/ioab242] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/15/2021] [Accepted: 12/24/2021] [Indexed: 12/30/2022] Open
Abstract
In vitro follicle development (IVFD) is an adequate model to obtain basic knowledge of folliculogenesis and provides a tool for ovarian toxicity screening. IVFD yielding competent oocytes may also offer an option for fertility and species preservation. To promote follicle growth and oocyte maturation in vitro, various culture systems are utilized for IVFD in rodents, domestic animals, wild animals, nonhuman primates, and humans. Follicle culture conditions have been improved by optimizing gonadotropin levels, regulatory factors, nutrient supplements, oxygen concentration, and culture matrices. This review summarizes quality assessment of oocytes generated from in vitro-developed antral follicles from the preantral stage, including oocyte epigenetic and genetic profile, cytoplasmic and nuclear maturation, preimplantation embryonic development following in vitro fertilization, as well as pregnancy and live offspring after embryo transfer. The limitations of oocyte quality evaluation following IVFD and the gaps in our knowledge of IVFD to support proper oocyte development are also discussed. The information may advance our understanding of the requirements for IVFD, with a goal of producing competent oocytes with genetic integrity to sustain embryonic development resulting in healthy offspring.
Collapse
Affiliation(s)
- Jing Xu
- Correspondence: Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR 97006, USA. Tel: +1 5033465411; Fax: +1 5033465585; E-mail:
| | - Mary B Zelinski
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA,Department of Obstetrics and Gynecology, School of Medicine, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
33
|
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
|
34
|
Generation of offspring-producing 3D ovarian organoids derived from female germline stem cells and their application in toxicological detection. Biomaterials 2021; 279:121213. [PMID: 34715637 DOI: 10.1016/j.biomaterials.2021.121213] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/23/2021] [Accepted: 10/20/2021] [Indexed: 12/21/2022]
Abstract
In vitro production of oocytes capable of producing offspring has exciting potential applications in reproductive medicine. Here, we generated and characterized an ovarian organoid model derived from female germline stem cells using a three-dimensional culture system. We show that this model generated normal offspring and detected drug toxicity. The ovarian organoids could produce oocytes and exhibited endocrine functions. Single-cell analysis of ovarian organoids identified six ovarian cell lineages, such as germ, granulosa and theca cells, and produced gene-expression signatures for each cell type. Investigation of the expression patterns of genes related to meiosis and gene ontogeny analysis for germ cell clusters showed that a germ cell population was maintained in the ovarian organoids. Moreover, flow cytometric analysis confirmed that the population of germ cells could be maintained on the organoids and showed that ascorbic acid treatment had a beneficial effect of germ cell population maintenance on the organoids. Furthermore, we demonstrated the successful production of offspring from oocytes derived from ovarian organoids. Finally, we showed the ovarian organoids had the potential to drug toxicological detection. For example, we found that salinomycin impaired the formation of ovarian organoids and germ cell population maintenance by inducing apoptosis. These results indicate that the female germline stem cell-derived ovarian organoids represent a valuable model system for generating oocytes that can yield offspring, and provide a novel model for drug screening and toxicological detection.
Collapse
|
35
|
Lee EB, Chakravarthi VP, Wolfe MW, Rumi MAK. ERβ Regulation of Gonadotropin Responses during Folliculogenesis. Int J Mol Sci 2021; 22:ijms221910348. [PMID: 34638689 PMCID: PMC8508937 DOI: 10.3390/ijms221910348] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 12/11/2022] Open
Abstract
Gonadotropins are essential for regulating ovarian development, steroidogenesis, and gametogenesis. While follicle stimulating hormone (FSH) promotes the development of ovarian follicles, luteinizing hormone (LH) regulates preovulatory maturation of oocytes, ovulation, and formation of corpus luteum. Cognate receptors of FSH and LH are G-protein coupled receptors that predominantly signal through cAMP-dependent and cAMP-independent mechanisms that activate protein kinases. Subsequent vital steps in response to gonadotropins are mediated through activation or inhibition of transcription factors required for follicular gene expression. Estrogen receptors, classical ligand-activated transcriptional regulators, play crucial roles in regulating gonadotropin secretion from the hypothalamic-pituitary axis as well as gonadotropin function in the target organs. In this review, we discuss the role of estrogen receptor β (ERβ) regulating gonadotropin response during folliculogenesis. Ovarian follicles in Erβ knockout (ErβKO) mutant female mice and rats cannot develop beyond the antral state, lack oocyte maturation, and fail to ovulate. Theca cells (TCs) in ovarian follicles express LH receptor, whereas granulosa cells (GCs) express both FSH receptor (FSHR) and LH receptor (LHCGR). As oocytes do not express the gonadotropin receptors, the somatic cells play a crucial role during gonadotropin induced oocyte maturation. Somatic cells also express high levels of estrogen receptors; while TCs express ERα and are involved in steroidogenesis, GCs express ERβ and are involved in both steroidogenesis and folliculogenesis. GCs are the primary site of ERβ-regulated gene expression. We observed that a subset of gonadotropin-induced genes in GCs, which are essential for ovarian follicle development, oocyte maturation and ovulation, are dependent on ERβ. Thus, ERβ plays a vital role in regulating the gonadotropin responses in ovary.
Collapse
Affiliation(s)
- Eun B. Lee
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (V.P.C.)
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | - V. Praveen Chakravarthi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (V.P.C.)
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA;
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Michael W. Wolfe
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA;
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - M. A. Karim Rumi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (E.B.L.); (V.P.C.)
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Correspondence: ; Tel.: +1-913-588-8059
| |
Collapse
|
36
|
Stejskalová A, Vankelecom H, Sourouni M, Ho MY, Götte M, Almquist BD. In vitro modelling of the physiological and diseased female reproductive system. Acta Biomater 2021; 132:288-312. [PMID: 33915315 DOI: 10.1016/j.actbio.2021.04.032] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 04/13/2021] [Accepted: 04/15/2021] [Indexed: 02/06/2023]
Abstract
The maladies affecting the female reproductive tract (FRT) range from infections to endometriosis to carcinomas. In vitro models of the FRT play an increasingly important role in both basic and translational research, since the anatomy and physiology of the FRT of humans and other primates differ significantly from most of the commonly used animal models, including rodents. Using organoid culture to study the FRT has overcome the longstanding hurdle of maintaining epithelial phenotype in culture. Both ECM-derived and engineered materials have proved critical for maintaining a physiological phenotype of FRT cells in vitro by providing the requisite 3D environment, ligands, and architecture. Advanced materials have also enabled the systematic study of factors contributing to the invasive metastatic processes. Meanwhile, microphysiological devices make it possible to incorporate physical signals such as flow and cyclic exposure to hormones. Going forward, advanced materials compatible with hormones and optimised to support FRT-derived cells' long-term growth, will play a key role in addressing the diverse array of FRT pathologies and lead to impactful new treatments that support the improvement of women's health. STATEMENT OF SIGNIFICANCE: The female reproductive system is a crucial component of the female anatomy. In addition to enabling reproduction, it has wide ranging influence on tissues throughout the body via endocrine signalling. This intrinsic role in regulating normal female biology makes it susceptible to a variety of female-specific diseases. However, the complexity and human-specific features of the reproductive system make it challenging to study. This has spurred the development of human-relevant in vitro models for helping to decipher the complex issues that can affect the reproductive system, including endometriosis, infection, and cancer. In this Review, we cover the current state of in vitro models for studying the female reproductive system, and the key role biomaterials play in enabling their development.
Collapse
|
37
|
Alaee S, Asadollahpour R, Hosseinzadeh Colagar A, Talaei-Khozani T. The decellularized ovary as a potential scaffold for maturation of preantral ovarian follicles of prepubertal mice. Syst Biol Reprod Med 2021; 67:413-427. [PMID: 34445905 DOI: 10.1080/19396368.2021.1968542] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
ABBREVIATIONS GAG: glycosaminoglycan; ECM: extracellular matrix; 2D: two-dimensional; E2: estradiol; P4: progesterone; BMP15: bone morphogenetic protein 15; GDF9: growth differentiation factor 9; ZP2: zona pellucida 2; Gdf9: growth/differentiation factor-9; Bmp6: bone morphogenetic protein 6; Bmp15: bone morphogenetic protein 15.
Collapse
Affiliation(s)
- Sanaz Alaee
- Department of Reproductive Biology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Raheleh Asadollahpour
- Department of Molecular and Cell Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar, Iran
| | | | - Tahereh Talaei-Khozani
- Tissue Engineering Lab, Department of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
38
|
Contrasting effects of the Toll-like receptor 4 in determining ovarian follicle endowment and fertility in female adult mice. ZYGOTE 2021; 30:227-233. [PMID: 34405787 DOI: 10.1017/s096719942100054x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Toll-like receptor 4 (TLR4) is best known for its role in bacteria-produced lipopolysaccharide recognition. Regarding female reproduction, TLR4 is expressed by murine cumulus cells and participates in ovulation and in cumulus-oocyte complex (COC) expansion, maternal-fetal interaction and preterm labour. Despite these facts, the role of TLR4 in ovarian physiology is not fully understood. Therefore, the aim of the present study was to investigate the effects of TLR4 genetic ablation on mice folliculogenesis and female fertility, through analysis of reproductive crosses, ovarian responsiveness and follicular quantification in TLR4-/- (n = 94) and C57BL/6 mice [wild type (WT), n = 102]. TLR4-deficient pairs showed a reduced number of pups per litter (P = 0.037) compared with WT. TLR4-/- mice presented more primordial, primary, secondary and antral follicles (P < 0.001), however there was no difference in estrous cyclicity (P > 0.05). A lower (P = 0.006) number of COC was recovered from TLR4-/- mice oviducts after superovulation, and in heterozygous pairs, TLR4-/- females also showed a reduction in the pregnancy rate and in the number of fetuses per uterus (P = 0.007) when compared with WT. Altogether, these data suggest that TLR4 plays a role in the regulation of murine folliculogenesis and in determining ovarian endowment. TLR4 deficiency may affect ovulation and pregnancy rates, potentially decreasing fertility, therefore the potential side effects of its blockade have to be carefully investigated.
Collapse
|
39
|
Xiang D, Liu Y, Zhou E, Wang Y. Advances in the applications of polymer biomaterials for in vitro follicle culture. Biomed Pharmacother 2021; 140:111422. [PMID: 34098195 DOI: 10.1016/j.biopha.2021.111422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 02/05/2021] [Accepted: 02/16/2021] [Indexed: 12/17/2022] Open
Abstract
The ovarian reserve (OR) indicates ovarian function by representing the quantity and quality of ovarian follicles, and it gradually decreases with increasing age. With the prolongation of women's lives, the protection provided by estrogen is lost for decades in postmenopausal women, and the related cardiovascular and cerebrovascular diseases, osteoporosis, and decreased immunity are the main risk factors affecting women's quality of life and longevity. Pharmacologic hormone replacement therapy (PHRT) has been controversial, and the construction of artificial ovary (AO) has attracted increasing attention. The most critical step of AO generation is the establishment of an in vitro culture (IVC) system to support the development of isolated follicles. This article mainly compares the advantages and disadvantages of different polymer biomaterials for use in follicle IVC, provides theoretical support for the development and construction of the follicle IVC system using natural biological materials, and provides a theoretical basis for establishing mature AO technology.
Collapse
Affiliation(s)
- Du Xiang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, 169 Donghu Road, Wuhan, Hubei 430071, China
| | - Yang Liu
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, 169 Donghu Road, Wuhan, Hubei 430071, China
| | - Encheng Zhou
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, 169 Donghu Road, Wuhan, Hubei 430071, China
| | - Yanfeng Wang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, 169 Donghu Road, Wuhan, Hubei 430071, China.
| |
Collapse
|
40
|
Chakravarthi VP, Ratri A, Masumi S, Borosha S, Ghosh S, Christenson LK, Roby KF, Wolfe MW, Rumi MAK. Granulosa cell genes that regulate ovarian follicle development beyond the antral stage: The role of estrogen receptor β. Mol Cell Endocrinol 2021; 528:111212. [PMID: 33676987 PMCID: PMC8916094 DOI: 10.1016/j.mce.2021.111212] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 02/06/2023]
Abstract
Follicle development beyond the preantral stage is dependent on gonadotropins. FSH signaling is crucial for the advancement of preantral follicles to the antral stage, and LH signaling is essential for further maturation of preovulatory follicles. Estrogen is intricately tied to gonadotropin signaling during the advanced stages of folliculogenesis. We observed that Erβnull ovarian follicles fail to develop beyond the antral stage, even after exogenous gonadotropin stimulation. As ERβ is primarily expressed in the granulosa cells (GCs), we explored the gonadotropin-regulated GC genes that induce maturation of antral follicles. Synchronized follicle development was induced by administration of exogenous gonadotropins to wildtype 4-wk-old female rats. The GC transcriptome was analyzed via RNA-sequencing before and after gonadotropin stimulation. An Erβnull mutant model that fails to show follicle maturation was also included in order to identify the ERβ-regulated genes involved at this step. We observed that specific groups of genes were differentially expressed in response to PMSG or hCG administration in wildtype rats. While some of the PMSG or hCG-induced genes showed a similar expression pattern in Erβnull GCs, a subset of PMSG- or hCG-induced genes showed a differential expression pattern in Erβnull GCs. These latter ERβ-regulated genes included previously known FSH or LH target genes including Lhcgr, Cyp11a1, Cyp19a1, Pgr, Runx2, Egfr, Kiss1, and Ptgs2, which are involved in follicle development, oocyte maturation, and ovulation. We also identified novel ERβ-regulated genes including Jaml, Galnt6, Znf750, Dusp9, Wnt16, and Mageb16 that failed to respond to gonadotropin stimulation in Erβnull GCs. Our findings indicate that the gonadotropin-induced spatiotemporal pattern of gene expression is essential for ovarian follicle maturation beyond the antral stage. However, expression of a subset of those gonadotropin-induced genes is dependent on transcriptional regulation by ERβ.
Collapse
Affiliation(s)
| | - Anamika Ratri
- Department of Molecular and Integrative Physiology, Kansas City, KS, USA
| | - Saeed Masumi
- Department of Pathology and Laboratory Medicine, Kansas City, KS, USA
| | - Shaon Borosha
- Department of Pathology and Laboratory Medicine, Kansas City, KS, USA
| | - Subhra Ghosh
- Department of Pathology and Laboratory Medicine, Kansas City, KS, USA
| | - Lane K Christenson
- Department of Molecular and Integrative Physiology, Kansas City, KS, USA; Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, KS, USA
| | - Katherine F Roby
- Department of Anatomy and Cell Biology, Kansas City, KS, USA; Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, KS, USA
| | - Michael W Wolfe
- Department of Molecular and Integrative Physiology, Kansas City, KS, USA; Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, KS, USA
| | - M A Karim Rumi
- Department of Pathology and Laboratory Medicine, Kansas City, KS, USA; Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
41
|
Leptin decreases apoptosis and promotes the activation of primordial follicles through the phosphatidylinositol-3-kinase/protein kinase B pathway in cultured ovine ovarian tissue. ZYGOTE 2021; 29:445-451. [PMID: 33906701 DOI: 10.1017/s0967199421000034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
This study evaluated the effects of leptin on primordial follicle survival and activation after in vitro culture of ovine ovarian tissue and if leptin acts through the phosphatidylinositol-3-kinase/protein kinase B (PI3K/Akt) pathway. Ovarian fragments were fixed for histology (fresh control) or cultured for 7 days in control medium (α-MEM+) alone or supplemented with leptin (1, 5, 10, 25 or 50 ng/ml). Follicle morphology, activation and apoptosis were analyzed. Next, the fragments were cultured in the medium that showed the best results in the absence or the presence of the PI3K inhibitor (LY294002), and immunohistostaining of p-Akt protein was assessed. After culture, the percentage of normal follicles decreased (P < 0.05) in all treatments compared with the fresh control. Moreover, control medium and 1 ng/ml leptin had similar (P > 0.05) percentages of normal follicles, which were significantly higher than those in other treatments. However, culture with 1 ng/ml leptin maintained apoptosis similarly (P > 0.05) to that of the fresh control and lower (P < 0.05) than that in α-MEM+. Leptin did not influence follicle activation (P > 0.05) compared with the control medium (α-MEM+). Culture in 1 ng/ml leptin with LY294002 decreased the normal follicles and increased apoptosis, inhibited follicle activation (P < 0.05), and reduced p-Akt immunostaining, compared with the medium containing 1 ng/ml leptin without PI3K inhibitor. In conclusion, leptin at 1 ng/ml reduces apoptosis and promotes the activation of primordial follicles compared with the fresh control after in vitro culture of ovine ovarian tissue possibly through the PI3K/Akt pathway.
Collapse
|
42
|
Telfer EE, Andersen CY. In vitro growth and maturation of primordial follicles and immature oocytes. Fertil Steril 2021; 115:1116-1125. [PMID: 33823993 DOI: 10.1016/j.fertnstert.2021.03.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/02/2021] [Accepted: 03/02/2021] [Indexed: 12/18/2022]
Abstract
Cryopreservation of ovarian tissue to preserve the fertility of girls and young women at high risk of sterility is now widely practiced. Pieces of cryopreserved ovarian cortex can be thawed and autografted to restore fertility, but because of the risks of reintroduction of the cancer, transplantation may not be possible for girls and women with blood-borne leukemias or cancers with a high risk of ovarian metastasis. Cryopreserved ovarian tissue contains mainly primordial follicles but also provides access to immature oocytes from small antral follicles, which may be matured in vitro to provide an additional source of mature oocytes. So in cases in which transplantation is contraindicated, fertility restoration could be safely achieved in the laboratory either by in vitro maturation (IVM) of oocytes aspirated from growing follicles or by the complete in vitro growth (IVG) and maturation (IVM) of primordial follicles to produce fertile metaphase II (MII) oocytes. The development of IVM and IVG methods to support all stages of oocytes available within ovarian tissue will maximize the potential for all patients undergoing fertility preservation.
Collapse
Affiliation(s)
- Evelyn E Telfer
- Institute of Cell Biology, The University of Edinburgh, Edinburgh EH8 8XE, Scotland.
| | - Claus Yding Andersen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, Faculty of Health and Medical Science, University of Copenhagen, DK-2100 Copenhagen, Denmark
| |
Collapse
|
43
|
Doherty CA, Diegmiller R, Kapasiawala M, Gavis ER, Shvartsman SY. Coupled oscillators coordinate collective germline growth. Dev Cell 2021; 56:860-870.e8. [PMID: 33689691 PMCID: PMC8265018 DOI: 10.1016/j.devcel.2021.02.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 12/03/2020] [Accepted: 02/16/2021] [Indexed: 12/20/2022]
Abstract
Developing oocytes need large supplies of macromolecules and organelles. A conserved strategy for accumulating these products is to pool resources of oocyte-associated germline nurse cells. In Drosophila, these cells grow more than 100-fold to boost their biosynthetic capacity. No previously known mechanism explains how nurse cells coordinate growth collectively. Here, we report a cell cycle-regulating mechanism that depends on bidirectional communication between the oocyte and nurse cells, revealing the oocyte as a critical regulator of germline cyst growth. Transcripts encoding the cyclin-dependent kinase inhibitor, Dacapo, are synthesized by the nurse cells and actively localized to the oocyte. Retrograde movement of the oocyte-synthesized Dacapo protein to the nurse cells generates a network of coupled oscillators that controls the cell cycle of the nurse cells to regulate cyst growth. We propose that bidirectional nurse cell-oocyte communication establishes a growth-sensing feedback mechanism that regulates the quantity of maternal resources loaded into the oocyte.
Collapse
Affiliation(s)
- Caroline A Doherty
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ 08540, USA
| | - Rocky Diegmiller
- Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ 08540, USA; Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08540, USA
| | - Manisha Kapasiawala
- Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ 08540, USA; Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08540, USA
| | - Elizabeth R Gavis
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540, USA.
| | - Stanislav Y Shvartsman
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540, USA; Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ 08540, USA; Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08540, USA; Center for Computational Biology, Flatiron Institute, New York, NY 10010, USA.
| |
Collapse
|
44
|
Abbassi L, El-Hayek S, Carvalho KF, Wang W, Yang Q, Granados-Aparici S, Mondadori R, Bordignon V, Clarke HJ. Epidermal growth factor receptor signaling uncouples germ cells from the somatic follicular compartment at ovulation. Nat Commun 2021; 12:1438. [PMID: 33664246 PMCID: PMC7933413 DOI: 10.1038/s41467-021-21644-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 02/07/2021] [Indexed: 01/31/2023] Open
Abstract
Germ cells are physically coupled to somatic support cells of the gonad during differentiation, but this coupling must be disrupted when they are mature, freeing them to participate in fertilization. In mammalian females, coupling occurs via specialized filopodia that project from the ovarian follicular granulosa cells to the oocyte. Here, we show that signaling through the epidermal growth factor receptor (EGFR) in the granulosa, which becomes activated at ovulation, uncouples the germ and somatic cells by triggering a massive and temporally synchronized retraction of the filopodia. Although EGFR signaling triggers meiotic maturation of the oocyte, filopodial retraction is independent of the germ cell state, being regulated solely within the somatic compartment, where it requires ERK-dependent calpain-mediated loss of filopodia-oocyte adhesion followed by Arp2/3-mediated filopodial shortening. By uncovering the mechanism regulating germ-soma uncoupling at ovulation, our results open a path to improving oocyte quality in human and animal reproduction.
Collapse
Affiliation(s)
- Laleh Abbassi
- Research Institute of the McGill University Health Centre, Montreal, Canada
- Division of Experimental Medicine, McGill University, Montreal, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Stephany El-Hayek
- Research Institute of the McGill University Health Centre, Montreal, Canada
- Department of Biology, McGill University, Montreal, Canada
- Centre for Arab Genomic Studies, Dubai, United Arab Emirates
| | - Karen Freire Carvalho
- Research Institute of the McGill University Health Centre, Montreal, Canada
- Division of Experimental Medicine, McGill University, Montreal, Canada
| | - Wusu Wang
- Research Institute of the McGill University Health Centre, Montreal, Canada
- College of Animal Science and Technology, Northwest A&F University, Shaanxi, PR China
| | - Qin Yang
- Research Institute of the McGill University Health Centre, Montreal, Canada
| | | | - Rafael Mondadori
- Department of Animal Science, McGill University, Montreal, Canada
| | - Vilceu Bordignon
- Department of Animal Science, McGill University, Montreal, Canada
| | - Hugh J Clarke
- Research Institute of the McGill University Health Centre, Montreal, Canada.
- Division of Experimental Medicine, McGill University, Montreal, Canada.
- Department of Biology, McGill University, Montreal, Canada.
- Department of Obstetrics and Gynecology, McGill University, Montreal, Canada.
| |
Collapse
|
45
|
El-Derany MO, Said RS, El-Demerdash E. Bone Marrow-Derived Mesenchymal Stem Cells Reverse Radiotherapy-Induced Premature Ovarian Failure: Emphasis on Signal Integration of TGF-β, Wnt/β-Catenin and Hippo Pathways. Stem Cell Rev Rep 2021; 17:1429-1445. [PMID: 33594662 DOI: 10.1007/s12015-021-10135-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2021] [Indexed: 11/29/2022]
Abstract
Radiotherapy is an indispensable cancer treatment approach. However, it is associated with hazardous consequences on multiple organs characterized by insidious worsening severity over time. This study aimed to examine the potential therapeutic effects of bone marrow mesenchymal stem cells (BM-MSCs) in radiation-induced premature ovarian failure (POF). Exposing female rats to 3.2 Gy whole-body ϒ-rays successfully induced POF. One week later, a single intravenous injection of BM-MSCs (2*106) cells was administered. BM-MSCs perfectly home to the damaged ovaries, enhanced ovarian follicle pool, and preserved the ovarian function manifested by restoring serum estradiol and follicle stimulating hormone levels, besides, rescuing the fertility outcomes of irradiated rats. These events have been associated with inhibiting ovarian apoptosis (Bax/Bcl2, caspase 3) and enhancing proliferation (PCNA). Interestingly, BM-MSCs reversed the inhibition of ovarian FOXO3 expression induced by radiation which resulted in increased primordial follicles stock. Moreover, BM-MSCs recovered the suppressed folliculogenesis process induced by radiation through upregulating FOXO1, GDF-9, and Fst genes expression accompanied by downregulating TGF-β which enhanced granulosa cells proliferation and secondary follicle development. Mechanistically, BM-MSCs miRNAs epigenetically upregulate Wnt/β-catenin and Hippo signaling pathways which are implicated in ovarian follicles growth and maturation. Therefore, BM-MSCs presented a ray of hope in the treatment of radiation-associated POF through genetic and epigenetic modulation of the integrated TGF-β, Wnt/β-catenin, and Hippo pathways which control apoptosis, proliferation, and differentiation of ovarian follicles.
Collapse
Affiliation(s)
- Marwa O El-Derany
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Riham S Said
- Department of Drug Radiation Research, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Ebtehal El-Demerdash
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt.
| |
Collapse
|
46
|
Zhang Y, Zhou X, Zhu Y, Wang H, Xu J, Su Y. Current mechanisms of primordial follicle activation and new strategies for fertility preservation. Mol Hum Reprod 2021; 27:6128515. [PMID: 33538812 DOI: 10.1093/molehr/gaab005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
Premature ovarian insufficiency (POI) is characterized by symptoms caused by ovarian dysfunction in patients aged <40 years. It is associated with a shortened reproductive lifespan. The only effective treatment for patients who are eager to become pregnant is IVF/Embryo Transfer (ET) using oocytes donated by young women. However, the use of the technique is constrained by the limited supply of oocytes and ethical issues. Some patients with POI still have some residual follicles in the ovarian cortex, which are not regulated by gonadotropin. These follicles are dormant. Therefore, activating dormant primordial follicles (PFs) to obtain high-quality oocytes for assisted reproductive technology may bring new hope for patients with POI. Therefore, this study aimed to explore the factors related to PF activation, such as the intercellular signaling network, the internal microenvironment of the ovary and the environment of the organism. In addition, we discussed new strategies for fertility preservation, such as in vitro activation and stem cell transplantation.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing 210004, China
| | - Xiaomei Zhou
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing 210004, China
| | - Ye Zhu
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing 210004, China
| | - Hanbin Wang
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing 210004, China
| | - Juan Xu
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing 210004, China
| | - Yiping Su
- Department of Gynecology, Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing 210004, China
| |
Collapse
|
47
|
Kehoe S, Jewgenow K, Johnston PR, Mbedi S, Braun BC. Signalling pathways and mechanistic cues highlighted by transcriptomic analysis of primordial, primary, and secondary ovarian follicles in domestic cat. Sci Rep 2021; 11:2683. [PMID: 33514822 PMCID: PMC7846758 DOI: 10.1038/s41598-021-82051-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 01/11/2021] [Indexed: 12/16/2022] Open
Abstract
In vitro growth (IVG) of dormant primordial ovarian follicles aims to produce mature competent oocytes for assisted reproduction. Success is dependent on optimal in vitro conditions complemented with an understanding of oocyte and ovarian follicle development in vivo. Complete IVG has not been achieved in any other mammalian species besides mice. Furthermore, ovarian folliculogenesis remains sparsely understood overall. Here, gene expression patterns were characterised by RNA-sequencing in primordial (PrF), primary (PF), and secondary (SF) ovarian follicles from Felis catus (domestic cat) ovaries. Two major transitions were investigated: PrF-PF and PF-SF. Transcriptional analysis revealed a higher proportion in gene expression changes during the PrF-PF transition. Key influencing factors during this transition included the interaction between the extracellular matrix (ECM) and matrix metalloproteinase (MMPs) along with nuclear components such as, histone HIST1H1T (H1.6). Conserved signalling factors and expression patterns previously described during mammalian ovarian folliculogenesis were observed. Species-specific features during domestic cat ovarian folliculogenesis were also found. The signalling pathway terms "PI3K-Akt", "transforming growth factor-β receptor", "ErbB", and "HIF-1" from the functional annotation analysis were studied. Some results highlighted mechanistic cues potentially involved in PrF development in the domestic cat. Overall, this study provides an insight into regulatory factors and pathways during preantral ovarian folliculogenesis in domestic cat.
Collapse
Affiliation(s)
- Shauna Kehoe
- Reproduction Biology Department, Leibniz Institute for Zoo and Wildlife Research, Alfred-Kowalke-Straße 17, 10315, Berlin, Germany.
| | - Katarina Jewgenow
- Reproduction Biology Department, Leibniz Institute for Zoo and Wildlife Research, Alfred-Kowalke-Straße 17, 10315, Berlin, Germany
| | - Paul R Johnston
- Berlin Center for Genomics in Biodiversity Research BeGenDiv, Königin-Luise-Straße 6-8, D-14195, Berlin, Germany
- Leibniz-Institute of Freshwater Ecology and Inland Fisheries, Müggelseedamm 310, 12587, Berlin, Germany
- Freie Universität Berlin, Institut für Biologie, Königin-Luise-Straße 1-3, 14195, Berlin, Germany
| | - Susan Mbedi
- Berlin Center for Genomics in Biodiversity Research BeGenDiv, Königin-Luise-Straße 6-8, D-14195, Berlin, Germany
- Museum für Naturkunde, Invalidenstraße 43, 10115, Berlin, Germany
| | - Beate C Braun
- Reproduction Biology Department, Leibniz Institute for Zoo and Wildlife Research, Alfred-Kowalke-Straße 17, 10315, Berlin, Germany
| |
Collapse
|
48
|
Kuyucu Y, Tap Ö. The effects of the anti-Müllerian hormone on folliculogenesis in rats: light and electron microscopic evaluation. Ultrastruct Pathol 2021; 45:59-70. [PMID: 33491534 DOI: 10.1080/01913123.2021.1877378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
In this study, we evaluated the effects of anti-Müllerian hormone on follicle development and oocyte quality with light and electron microscopy. Twenty-four adult female rats were divided into four groups. After estrous cycle synchronization, on the first day, control group rats were injected with 0.5 ml saline, 2nd, 3rd, and 4th groups were injected 1 µgr, 2 µgr, and 5 µgr anti-Müllerian hormone, respectively. On the third day, intracardiac blood samples were taken for follicle-stimulating hormone, luteinizing hormone, estradiol, and progesterone serum level measurements. Ovaries were obtained for light and electron microscopic examinations. Secondary (antral) follicles were decreased while atretic follicles were increased in number parallel with an increased dose of anti-Müllerian hormone injection. Atresia of the follicles was demonstrated with apoptosis of granulosa cells characterized by apoptotic bodies and with paraptosis characterized by the vacuole formation in the cytoplasm, enlargement of granular endoplasmic reticulum cisternae and perinuclear cisternae in granulosa cells. Premature luteinization characterized by increased lipid droplets, mitochondria with tubular cristae, and smooth-surfaced endoplasmic reticulum in the cytoplasm of granulosa cells were detected in some growing follicles. In the anti-Müllerian hormone injected experimental groups, cystic follicles characterized by a large antrum, attenuated granulosa cell layer, and flattened granulosa cells that face the antrum were observed. Corpus luteum and stroma were similar in all groups. It was concluded that increasing doses of anti-Müllerian hormone caused increased atresia in developing follicles, premature luteinization of granulosa cells in some follicles, and cystic follicle formation in the further developing follicles.
Collapse
Affiliation(s)
- Yurdun Kuyucu
- Department of Histology and Embryology, Çukurova University, Adana, Turkey
| | - Özgül Tap
- Department of Histology and Embryology, Çukurova University, Adana, Turkey
| |
Collapse
|
49
|
Adu-Gyamfi EA, Czika A, Liu TH, Gorleku PN, Fondjo LA, Djankpa FT, Ding YB, Wang YX. Ephrin and Eph receptor signaling in female reproductive physiology and pathology†. Biol Reprod 2020; 104:71-82. [PMID: 32940657 DOI: 10.1093/biolre/ioaa171] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/19/2020] [Accepted: 09/13/2020] [Indexed: 12/12/2022] Open
Abstract
Ephrins are ligands of Eph receptors (Ephs); both of which are sorted into two classes, A and B. There are five types of ephrin-As (ephrin-A1-5) and three types of ephrin-Bs (ephrin-B1-3). Also, there are 10 types of EphAs (EphA1-10) and six types of EphBs (EphB1-6). Binding of ephrins to the Eph receptors activates signaling cascades that regulate several biological processes such as cellular proliferation, differentiation, migration, angiogenesis, and vascular remodeling. Clarification of their roles in the female reproductive system is crucial to understanding the physiology and pathology of this system. Such knowledge will also create awareness regarding the importance of these molecules in diagnostic, prognostic, and therapeutic medicine. Hence, we have discussed the involvement of these molecules in the physiological and pathological events that occur within the female reproductive system. The evidence so far suggests that the ephrins and the Eph receptors modulate folliculogenesis, ovulation, embryo transport, implantation, and placentation. Abnormal expression of some of these molecules is associated with polycystic ovarian syndrome, ovarian cancer, tubal pregnancy, endometrial cancer, uterine leiomyoma (fibroids), cervical cancer, and preeclampsia, suggesting the need to utilize these molecules in the clinical setting. To enhance a quick development of this gradually emerging field in female reproductive medicine, we have highlighted some "gaps in knowledge" that need prospective investigation.
Collapse
Affiliation(s)
- Enoch Appiah Adu-Gyamfi
- Department of Reproductive Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing, People's Republic of China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, People's Republic of China
| | - Armin Czika
- Department of Reproductive Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing, People's Republic of China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, People's Republic of China
| | - Tai-Hang Liu
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, People's Republic of China
| | - Philip Narteh Gorleku
- Department of Medical Imaging, School of Medical Sciences, University of Cape Coast, Cape Coast, Republic of Ghana
| | - Linda Ahenkorah Fondjo
- Department of Molecular Medicine, School of Medicine and Dentistry, Kwame Nkrumah University of Science and Technology, Kumasi, Republic of Ghana
| | - Francis Tanam Djankpa
- Department of Physiology, School of Medical Sciences, University of Cape Coast, Cape Coast, Republic of Ghana
| | - Yu-Bin Ding
- Department of Reproductive Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing, People's Republic of China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, People's Republic of China
| | - Ying-Xiong Wang
- Department of Reproductive Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing, People's Republic of China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, People's Republic of China
| |
Collapse
|
50
|
Mastrorocco A, Cacopardo L, Martino NA, Fanelli D, Camillo F, Ciani E, Roelen BAJ, Ahluwalia A, Dell’Aquila ME. One-step automated bioprinting-based method for cumulus-oocyte complex microencapsulation for 3D in vitro maturation. PLoS One 2020; 15:e0238812. [PMID: 32915922 PMCID: PMC7485809 DOI: 10.1371/journal.pone.0238812] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 08/23/2020] [Indexed: 12/14/2022] Open
Abstract
Three-dimensional in vitro maturation (3D IVM) is a promising approach to improve IVM efficiency as it could prevent cumulus-oocyte complex (COC) flattening and preserve its structural and functional integrity. Methods reported to date have low reproducibility and validation studies are limited. In this study, a bioprinting based production process for generating microbeads containing a COC (COC-microbeads) was optimized and its validity tested in a large animal model (sheep). Alginate microbeads were produced and characterized for size, shape and stability under culture conditions. COC encapsulation had high efficiency and reproducibility and cumulus integrity was preserved. COC-microbeads underwent IVM, with COCs cultured in standard 2D IVM as controls. After IVM, oocytes were analyzed for nuclear chromatin configuration, bioenergetic/oxidative status and transcriptional activity of genes biomarker of mitochondrial activity (TFAM, ATP6, ATP8) and oocyte developmental competence (KHDC3, NLRP5, OOEP and TLE6). The 3D system supported oocyte nuclear maturation more efficiently than the 2D control (P<0.05). Ooplasmic mitochondrial activity and reactive oxygen species (ROS) generation ability were increased (P<0.05). Up-regulation of TFAM, ATP6 and ATP8 and down-regulation of KHDC3, NLRP5 expression were observed in 3D IVM. In conclusion, the new bioprinting method for producing COC-microbeads has high reproducibility and efficiency. Moreover, 3D IVM improves oocyte nuclear maturation and relevant parameters of oocyte cytoplasmic maturation and could be used for clinical and toxicological applications.
Collapse
Affiliation(s)
- Antonella Mastrorocco
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
- * E-mail:
| | | | - Nicola Antonio Martino
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
| | - Diana Fanelli
- Department of Veterinary Sciences, University of Pisa, Pisa, Italy
| | | | - Elena Ciani
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
| | - Bernard A. J. Roelen
- Department of Clinical Sciences, Embryology, Anatomy and Physiology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Arti Ahluwalia
- Research Centre E. Piaggio, University of Pisa, Pisa, Italy
- Department of Information Engineering, University of Pisa, Pisa, Italy
| | - Maria Elena Dell’Aquila
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|