1
|
Shafiei G, Talaei SA, Enderami SE, Mahabady MK, Mahabadi JA. Pluripotent stem cell-derived gametes: A gap for infertility treatment and reproductive medicine in the future. Tissue Cell 2025; 95:102904. [PMID: 40203683 DOI: 10.1016/j.tice.2025.102904] [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: 09/30/2024] [Revised: 03/26/2025] [Accepted: 03/29/2025] [Indexed: 04/11/2025]
Abstract
Infertility affects 10-15 % of reproductive-age couples worldwide, with male infertility linked to sperm dysfunction and female infertility caused by ovulation disorders and reproductive abnormalities. Stem cell research presents a promising avenue for infertility treatment through germ cell differentiation. However, standardizing differentiation protocols and ensuring the functionality of in vitro-derived gametes remain significant challenges before clinical application becomes feasible.
Collapse
Affiliation(s)
- Golnaz Shafiei
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Sayyed Alireza Talaei
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Ehsan Enderami
- Immunogenetics Research Center, Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mahmood Khaksary Mahabady
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Javad Amini Mahabadi
- Gametogenesis Research Center, Kashan University of Medical Science, Kashan, Iran.
| |
Collapse
|
2
|
Johnson J, Emerson JW, Smith A, Medina K, Telfer EE, Anderson RA, Lawley SD. Modeling the extension of ovarian function after therapeutic targeting of the primordial follicle reserve. Hum Reprod Update 2025:dmaf009. [PMID: 40324778 DOI: 10.1093/humupd/dmaf009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 02/20/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Women are increasingly choosing to delay childbirth, and those with low ovarian reserves indicative of primary ovarian insufficiency are at risk for sub- and infertility and also the early onset of menopause. Experimental strategies that promise to extend the duration of ovarian function in women are currently being developed. One strategy is to slow the rate of loss of existing primordial follicles (PFs), and a second is to increase, or 'boost', the number of autologous PFs in the human ovary. In both cases, the duration of ovarian function would be expected to be lengthened, and menopause would be delayed. This might be accompanied by an extended production of mature oocytes of sufficient quality to extend the fertile lifespan. OBJECTIVE AND RATIONALE In this work, we consider how slowing physiological ovarian aging might improve the health and well-being of patients, and summarize the current state-of-the-art of approaches being developed. We then use mathematical modeling to determine how interventions are likely to influence the duration of ovarian function quantitatively. Finally, we consider efficacy benchmarks that should be achieved so that individuals will benefit, and propose criteria that could be used to monitor ongoing efficacy in different patients as these strategies are being validated. SEARCH METHODS Current methods to estimate the size of the ovarian reserve and its relationship to the timing of the menopausal transition and menopause were compiled, and publications establishing methods designed to slow loss of the ovarian reserve or to deliver additional ovarian PFs to patients were identified. OUTCOMES We review our current understanding of the consequences of reproductive aging in women, and compare different approaches that may extend ovarian function in women at risk for POI. We also provide modeling of primordial reserve decay in the presence of therapies that slow PF loss or boost PF numbers. An interactive online tool is provided that estimates how different interventions would impact the duration of ovarian function across the natural population. Modeling output shows that treatments that slow PF loss would need to be applied as early as possible and for many years to achieve significant delay of menopause. In contrast, treatments that add additional PFs should occur as late as possible relative to the onset of menopause. Combined approaches slowing ovarian reserve loss while also boosting numbers of (new) PFs would likely offer some additional benefits in delaying menopause. WIDER IMPLICATIONS Extending ovarian function, and perhaps the fertile lifespan, is on the horizon for at least some patients. Modeling ovarian aging with and without such interventions complements and helps guide the clinical approaches that will achieve this goal. REGISTRATION NUMBER Not applicable.
Collapse
Affiliation(s)
- Joshua Johnson
- Division of Reproductive Sciences, Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Colorado Denver (AMC), Aurora, CO, USA
| | - John W Emerson
- Department of Statistics and Data Science, Yale University, New Haven, CT, USA
| | - Annika Smith
- Division of Reproductive Sciences, Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Colorado Denver (AMC), Aurora, CO, USA
| | - Kayla Medina
- Division of Reproductive Sciences, Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Colorado Denver (AMC), Aurora, CO, USA
| | - Evelyn E Telfer
- Institute of Cell Biology, Hugh Robson Building, University of Edinburgh, Edinburgh, UK
- Centre for Reproductive Health, Institute of Regeneration and Repair, Edinburgh, UK
| | - Richard A Anderson
- Centre for Reproductive Health, Institute of Regeneration and Repair, Edinburgh, UK
| | - Sean D Lawley
- Department of Mathematics, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
3
|
Sikiru AB, Truong MN, Zohdy W. Future prospects for the advancement of treatment of men with NOA: focus on gene editing, artificial sperm, stem cells, and use of imaging. Asian J Androl 2025; 27:433-439. [PMID: 39422616 DOI: 10.4103/aja202486] [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: 01/27/2024] [Accepted: 08/20/2024] [Indexed: 10/19/2024] Open
Abstract
ABSTRACT Nonobstructive azoospermia (NOA) affects about 60% of men with azoospermia, representing a severe form of male infertility. The current approach to manage NOA primarily involves testicular sperm retrieval methods such as conventional testicular sperm extraction (c-TESE) and microdissection testicular sperm extraction (micro-TESE). While combining testicular sperm retrieval with intracytoplasmic sperm injection (ICSI) offers hope for patients, the overall sperm retrieval rate (SRR) stands at around 50%. In cases where micro-TESE fails to retrieve sperm, limited options, like donor sperm or adoption, can be problematic in certain cultural contexts. This paper delves into prospective treatments for NOA management. Gene editing technologies, particularly clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated (Cas) protein 9 (CRISPR/Cas9), hold potential for correcting genetic mutations underlying testicular dysfunction. However, these technologies face challenges due to their complexity, potential off-target effects, ethical concerns, and affordability. This calls for research to address key challenges associated with NOA management within the clinical settings. This also necessitate ongoing research essential for developing more sensitive diagnostic tests, validating novel treatments, and customizing current treatment strategies for individual patients. This review concluded that the future of NOA management may entail a combination of these treatment options, tailored to each patient's unique circumstances, providing a comprehensive approach to address NOA challenges.
Collapse
Affiliation(s)
- Akeem Babatunde Sikiru
- Department of Animal Science, Federal University of Agriculture Zuru (FUAZ), Kebbi 872101, Nigeria
| | - Manh Nguyen Truong
- Fertility Centre, Hanh Phuc International Hospital, Ho Chi Minh City 70000, Vietnam
| | - Wael Zohdy
- Andrology and STIs, Cairo University, Giza 12613, Egypt
| |
Collapse
|
4
|
Bhattacharya I, Nalinan LK, Anusree KV, Saleel A, Khamamkar A, Dey S. Evolving Lessons on Metazoan Primordial Germ Cells in Diversity and Development. Mol Reprod Dev 2025; 92:e70027. [PMID: 40349219 PMCID: PMC12066098 DOI: 10.1002/mrd.70027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 04/15/2025] [Indexed: 05/14/2025]
Abstract
Germ cells are pivotal for the continuation of biological species. The metazoan germline develops from primordial germ cells (PGCs) that undergo multiple rounds of mitotic divisions. The PGCs are specified by either maternal inheritance of asymmetrically polarized cytoplasmic mRNAs/proteins (found in roundworms, flies, fishes, frogs, and fowl) or via direct induction of epiblast cells from adjacent extraembryonic ectoderm in mammals. In all vertebrates, PGCs remain uncommitted to meiosis and migrate to colonize the developing gonadal ridge before sex determination. Multiple RNA-binding proteins (e.g., Vasa, Dnd, Dazl, etc.) play crucial roles in PGC identity, expansion, survival, and migration. Postsex determination in mouse embryos, Gata4, expressing nascent gonads, induces Dazl expression in newly arriving germ cells that supports retinoic acid-mediated induction of meiotic onset. This article briefly discusses the developmental events regulating the PGC specification and commitment in metazoans. We also highlight the recent progress towards the in vitro generation of functional PGC-like cells in rodents and humans.
Collapse
Affiliation(s)
- Indrashis Bhattacharya
- Department of ZoologyThe Central University of KeralaTejaswini Hills, Periye (PO)Kasaragod (DT)KeralaIndia
| | - Lakshmi K. Nalinan
- Department of ZoologyThe Central University of KeralaTejaswini Hills, Periye (PO)Kasaragod (DT)KeralaIndia
| | - K. V. Anusree
- Department of ZoologyThe Central University of KeralaTejaswini Hills, Periye (PO)Kasaragod (DT)KeralaIndia
| | - Ahmed Saleel
- Department of ZoologyThe Central University of KeralaTejaswini Hills, Periye (PO)Kasaragod (DT)KeralaIndia
| | - Aditi Khamamkar
- Manipal Centre for Biotherapeutics ResearchManipal Academy of Higher EducationManipalKarnatakaIndia
| | - Souvik Dey
- Manipal Centre for Biotherapeutics ResearchManipal Academy of Higher EducationManipalKarnatakaIndia
| |
Collapse
|
5
|
Wang X, Huang R, Liu L, Wang X, Zhang X. Evaluation and preservation of fertility in patients with hematologic malignancies. Cancer Lett 2025; 616:217569. [PMID: 39983893 DOI: 10.1016/j.canlet.2025.217569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 02/23/2025]
Abstract
For patients with hematologic malignancies, novel therapeutic strategies offer the potential to achieve a complete clinical response and long-term survival. However, declining fertility has become a significant concern, impacting long-term quality of life. Conventional high-dose chemotherapy and radiotherapy are known to reduce fertility or cause sterility. Moreover, limited clinical data are available on the effects of newer therapies, such as targeted treatments and chimeric antigen receptor (CAR)-T cell therapy, on fertility. Additionally, there is no standard method for preserving fertility in these patients. Male patients can opt for sperm cryopreservation, whereas female patients may preserve fertility through embryo, oocyte, or ovarian tissue cryopreservation. However, preserving fertility in prepubescent patients remains particularly challenging. Therefore, hematologists must educate patients about the potential gonadal toxicity of cancer treatments and offer the most appropriate fertility preservation options.
Collapse
Affiliation(s)
- Xiang Wang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China; Institute of Science Innovation for Blood Ecology and Intelligent Cells, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China; Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, China; State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400037, China; Jinfeng Laboratory, Chongqing, 400037, China
| | - Ruihao Huang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China; Institute of Science Innovation for Blood Ecology and Intelligent Cells, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China; Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, China; State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400037, China
| | - Lei Liu
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China; Institute of Science Innovation for Blood Ecology and Intelligent Cells, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China; Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, China; State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400037, China; Jinfeng Laboratory, Chongqing, 400037, China
| | - Xiaoqi Wang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China; Institute of Science Innovation for Blood Ecology and Intelligent Cells, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China; Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, China; State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400037, China.
| | - Xi Zhang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China; Institute of Science Innovation for Blood Ecology and Intelligent Cells, Xinqiao Hospital of Army Medical University, Chongqing, 400037, China; Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, 400037, China; State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing, 400037, China; Jinfeng Laboratory, Chongqing, 400037, China.
| |
Collapse
|
6
|
Novak BJ, Brand S, Phelan R, Plichta S, Ryder OA, Wiese RJ. Towards Practical Conservation Cloning: Understanding the Dichotomy Between the Histories of Commercial and Conservation Cloning. Animals (Basel) 2025; 15:989. [PMID: 40218382 PMCID: PMC11988126 DOI: 10.3390/ani15070989] [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: 01/31/2025] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Over 40 years ago, scientists imagined ways cloning could aid conservation of threatened taxa. The cloning of Dolly the sheep from adult somatic cells in 1996 was the breakthrough that finally enabled the conservation potential of the technology. Until the 2020s, conservation cloning research efforts yielded no management applications, leading many to believe cloning is not yet an effective conservation tool. In strong contrast, domestic taxa are cloned routinely for scientific and commercial purposes. In this review, we sought to understand the reasons for these divergent trends. We scoured peer-reviewed and gray literature and sent direct inquiries to scientists to analyze a more comprehensive history of the field than was analyzed in previous reviews. While most previous reviewers concluded that a lack of reproductive knowledge of wildlife species has hindered advances for wider conservation applications, we found that resource limitations (e.g., numbers of surrogates, sustainable funding) and widely held misconceptions about cloning are significant contributors to the stagnation of the field. Recent successes in cloning programs for the endangered black-footed ferret (Mustela nigripes) and Przewalski's horse (Equus przewalskii), the world's first true applied-conservation cloning efforts, are demonstrating that cloning can be used for significant conservation impact in the present. When viewed alongside the long history of cloning achievements, these programs emphasize the value of investing in the science and resources needed to meaningfully integrate cloning into conservation management, especially for species with limited genetic diversity that rely on the maintenance of small populations for many generations while conservationists work to restore habitat and mitigate threats in the wild.
Collapse
Affiliation(s)
- Ben J. Novak
- Revive & Restore, 1505 Bridgeway #203, Sausalito, CA 94965, USA
| | - Stewart Brand
- Revive & Restore, 1505 Bridgeway #203, Sausalito, CA 94965, USA
| | - Ryan Phelan
- Revive & Restore, 1505 Bridgeway #203, Sausalito, CA 94965, USA
| | - Sasha Plichta
- Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Oliver A. Ryder
- Beckman Center for Conservation Research, San Diego Zoo Wildlife Alliance, Escondido, CA 92027, USA
| | - Robert J. Wiese
- North Carolina Museum of Natural Sciences, Raleigh, NC 27601, USA;
| |
Collapse
|
7
|
Yu H, Wang Z, Ma J, Wang R, Yao S, Gu Z, Lin K, Li J, Young RS, Yu Y, Yu Y, Jin M, Chen D. The establishment and regulation of human germ cell lineage. Stem Cell Res Ther 2025; 16:139. [PMID: 40102947 PMCID: PMC11921702 DOI: 10.1186/s13287-025-04171-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 01/23/2025] [Indexed: 03/20/2025] Open
Abstract
The specification of primordial germ cells (PGCs) during early embryogenesis initiates the development of the germ cell lineage that ensures the perpetuation of genetic and epigenetic information from parents to offspring. Defects in germ cell development may lead to infertility or birth defects. Historically, our understanding of human PGCs (hPGCs) regulation has primarily been derived from studies in mice, given the ethical restrictions and practical limitations of human embryos at the stage of PGC specification. However, recent studies have increasingly highlighted significant mechanistic differences for PGC development in humans and mice. The past decade has witnessed the establishment of human pluripotent stem cell (hPSC)-derived hPGC-like cells (hPGCLCs) as new models for studying hPGC fate specification and differentiation. In this review, we systematically summarize the current hPSC-derived models for hPGCLC induction, and how these studies uncover the regulatory machinery for human germ cell fate specification and differentiation, forming the basis for reconstituting gametogenesis in vitro from hPSCs for clinical applications and disease modeling.
Collapse
Affiliation(s)
- Honglin Yu
- Center for Reproductive Medicine of The Second Affiliated Hospital, Center for Regeneration and Cell Therapy of Zhejiang, University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, 314400, Zhejiang, China
| | - Ziqi Wang
- Center for Reproductive Medicine of The Second Affiliated Hospital, Center for Regeneration and Cell Therapy of Zhejiang, University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, 314400, Zhejiang, China
| | - Jiayue Ma
- Center for Reproductive Medicine of The Second Affiliated Hospital, Center for Regeneration and Cell Therapy of Zhejiang, University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, 314400, Zhejiang, China
| | - Ruoming Wang
- Center for Reproductive Medicine of The Second Affiliated Hospital, Center for Regeneration and Cell Therapy of Zhejiang, University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, 314400, Zhejiang, China
| | - Shuo Yao
- Center for Reproductive Medicine of The Second Affiliated Hospital, Center for Regeneration and Cell Therapy of Zhejiang, University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, 314400, Zhejiang, China
| | - Zhaoyu Gu
- Center for Reproductive Medicine of The Second Affiliated Hospital, Center for Regeneration and Cell Therapy of Zhejiang, University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, 314400, Zhejiang, China
| | - Kexin Lin
- Center for Reproductive Medicine of The Second Affiliated Hospital, Center for Regeneration and Cell Therapy of Zhejiang, University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, 314400, Zhejiang, China
| | - Jinlan Li
- College of Animal & Veterinary Sciences, Southwest Minzu University, Chengdu, 610041, Sichuan, China
| | - Robert S Young
- Center for Global Health Research, Usher Institute, University of Edinburgh, 5-7 Little France Road, Edinburgh, EH16 4UX, UK
- Zhejiang University - University of Edinburgh Institute, Zhejiang University, Haining, 314400, Zhejiang, China
| | - Ya Yu
- Center for Reproductive Medicine of The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
| | - You Yu
- Center for Infection Immunity, Cancer of Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China.
| | - Min Jin
- Center for Reproductive Medicine of The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China.
| | - Di Chen
- Center for Reproductive Medicine of The Second Affiliated Hospital, Center for Regeneration and Cell Therapy of Zhejiang, University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, 314400, Zhejiang, China.
- State Key Laboratory of Biobased Transportation Fuel Technology, Haining, 314400, Zhejiang, China.
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
8
|
Yoshida T, Tsukamoto M, Kimura K, Tanaka M, Kuwamura M, Hatoya S. Establishment of feline embryonic stem cells from the inner cell mass of blastocysts produced in vitro. Regen Ther 2025; 28:63-72. [PMID: 39697661 PMCID: PMC11652941 DOI: 10.1016/j.reth.2024.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/12/2024] [Accepted: 11/17/2024] [Indexed: 12/20/2024] Open
Abstract
Introduction The rising number of cats as pets and the growing interest in animal welfare have led to an increased need for the latest treatments in feline veterinary medicine. Among these, veterinary regenerative medicine using pluripotent stem cells is gaining significant attention. However, there have been no reports on establishing feline embryonic stem cell (ESC) lines that possess the pluripotent potential and the ability to differentiate into three germ layers. Methods In this study, we isolated three inner cell masses from feline in vitro-derived blastocysts and subcultured them in a chemically defined medium (StemFit AK02N). We assessed the expression of undifferentiated markers, the ability to differentiate into the three germ layers, and the karyotype structure. Results We established three feline ESC lines. Feline ESCs exhibited positive staining for alkaline phosphatase. RT-qPCR analysis revealed that these cells express undifferentiated marker genes in vitro. Immunostaining and flow cytometry analysis demonstrated that feline ESCs express undifferentiated marker proteins in vitro. In the KSR/FBS medium with or without Activin A, feline ESCs differentiated into all three germ layers (ectoderm, endoderm, and mesoderm), expressing specific marker genes and proteins for each germ layer, as evidenced by RT-qPCR, immunostaining, and flow cytometry. Furthermore, we confirmed that feline ESCs formed teratomas comprising all three germ layers in mouse testes, demonstrating de novo pluripotency in vivo. We also verified that the feline ESCs maintained a normal karyotype. Conclusions We successfully established three feline ESC lines, each possessing pluripotent potential and capable of differentiating into all three germ layers, derived from the inner cell masses of blastocysts produced in vitro.
Collapse
Affiliation(s)
- Takumi Yoshida
- Department of Advanced Pathobiology, Graduate School of Veterinary Science, Osaka Metropolitan University, Izumisano, Osaka 598-8531, Japan
| | - Masaya Tsukamoto
- Department of Advanced Pathobiology, Graduate School of Veterinary Science, Osaka Metropolitan University, Izumisano, Osaka 598-8531, Japan
| | - Kazuto Kimura
- Department of Advanced Pathobiology, Graduate School of Veterinary Science, Osaka Metropolitan University, Izumisano, Osaka 598-8531, Japan
| | - Miyuu Tanaka
- Department of Integrated Structural Biosciences, Graduate School of Veterinary Science, Osaka Metropolitan University, Izumisano, Osaka 598-8531, Japan
| | - Mitsuru Kuwamura
- Department of Integrated Structural Biosciences, Graduate School of Veterinary Science, Osaka Metropolitan University, Izumisano, Osaka 598-8531, Japan
| | - Shingo Hatoya
- Department of Advanced Pathobiology, Graduate School of Veterinary Science, Osaka Metropolitan University, Izumisano, Osaka 598-8531, Japan
| |
Collapse
|
9
|
Wu B, Neupane J, Zhou Y, Zhang J, Chen Y, Surani MA, Zhang Y, Bao S, Li X. Stem cell-based embryo models: a tool to study early human development. SCIENCE CHINA. LIFE SCIENCES 2025:10.1007/s11427-024-2741-1. [PMID: 39969747 DOI: 10.1007/s11427-024-2741-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 09/23/2024] [Indexed: 02/20/2025]
Abstract
How a mammalian fertilized egg acquires totipotency and develops into a full-term offspring is a fundamental scientific question. Human embryonic development is difficult to study due to limited resources, technical challenges and ethics. Moreover, the precise regulatory mechanism underlying early human embryonic development remains unknown. In recent years, the emergence of stem cell-based embryo models (SCBEM) provides the opportunity to reconstitute pre- to post-implantation development in vitro. These models to some extent mimic the embryo morphologically and transcriptionally, and thus may be used to study key events in mammalian pre- and post-implantation development. Many groups have successfully generated SCBEM of the mouse and human. Here, we provide a comparative review of the mouse and human SCBEM, discuss the capability of these models to mimic natural embryos and give a perspective on their potential future applications.
Collapse
Affiliation(s)
- Baojiang Wu
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China
| | - Jitesh Neupane
- The Gurdon Institute, University of Cambridge, Cambridge, CB2 1QN, UK
| | - Yang Zhou
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China
| | - Jingcheng Zhang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Yanglin Chen
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China
| | - M Azim Surani
- The Gurdon Institute, University of Cambridge, Cambridge, CB2 1QN, UK
| | - Yong Zhang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China.
| | - Siqin Bao
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China.
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China.
| | - Xihe Li
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China.
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China.
- Inner Mongolia Saikexing Institute of Breeding and Reproductive Biotechnology in Domestic Animals, Hohhot, 011517, China.
| |
Collapse
|
10
|
Mu C, Li X, Yang J, Tian GG, Bai H, Lin W, Wang L, Wu J. Spatial Transcriptome and Single Nucleus Transcriptome Sequencing Reveals Tetrahydroxy Stilbene Glucoside Promotes Ovarian Organoids Development Through the Vegfa-Ephb2 Pair. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410098. [PMID: 39629971 PMCID: PMC11775562 DOI: 10.1002/advs.202410098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/03/2024] [Indexed: 01/30/2025]
Abstract
Ovarian dysfunction is a major factor leading to female infertility. Understanding how to improve or reshape ovarian function has become an important entry point for preventing and treating female infertility caused by ovarian dysfunction. Here, plant-derived compounds are screened for in vitro activity upon ovarian organoids derived from feeder-free female germline stem cells. Tetrahydroxy stilbene glucoside (TSG) is found to promote the development of ovarian organoids. Single nucleus transcriptome sequencing and spatial transcriptome sequencing are used to establish a comprehensive spatiotemporal map to elucidate the role of TSG in ovarian organoid development, encompassing cell types and subtypes, transcription factors, pseudo-time sequence, and cell communication dynamics. This analysis indicates that TSG promotes ovarian organoid development through the vascular endothelial growth factor A-Eph receptor B2 ligand-receptor pair between granulosa cells and oocytes. This study has enhanced the understanding of the mechanisms of ovarian organoid development, establishes a technical platform for screening compounds for treating infertility and related diseases, and lays a foundation for clinically applying plant-derived compounds.
Collapse
Affiliation(s)
- Chunlan Mu
- School of Basic Medical SciencesKey Laboratory of Fertility Preservation and Maintenance of Ministry of EducationNingxia Medical UniversityYinchuan750004China
- Key Laboratory for the Genetics of Developmental & Neuropsychiatric Disorders (Ministry of Education)Bio‐X InstitutesShanghai Jiao Tong UniversityShanghai200240China
| | - Xiaoyong Li
- Key Laboratory for the Genetics of Developmental & Neuropsychiatric Disorders (Ministry of Education)Bio‐X InstitutesShanghai Jiao Tong UniversityShanghai200240China
| | - Jiamei Yang
- School of Basic Medical SciencesKey Laboratory of Fertility Preservation and Maintenance of Ministry of EducationNingxia Medical UniversityYinchuan750004China
- School of PharmacologyNingxia Medical UniversityYinchuan750004China
| | - Geng G. Tian
- School of Agriculture and BiologyShanghai Jiao Tong UniversityShanghai200240China
| | - Hepeng Bai
- Key Laboratory for the Genetics of Developmental & Neuropsychiatric Disorders (Ministry of Education)Bio‐X InstitutesShanghai Jiao Tong UniversityShanghai200240China
| | - Wenhui Lin
- School of Life Sciences and BiotechnologyJoint International Research Laboratory of Metabolic and Developmental SciencesShanghai Jiao Tong UniversityShanghai200240China
| | - Linhui Wang
- Department of Urology, Changhai HospitalSecond Military Medical University (Naval Medical University)Shanghai200433China
- Shanghai Key Laboratory of Cell EngineeringShanghai200433China
| | - Ji Wu
- School of Basic Medical SciencesKey Laboratory of Fertility Preservation and Maintenance of Ministry of EducationNingxia Medical UniversityYinchuan750004China
- Key Laboratory for the Genetics of Developmental & Neuropsychiatric Disorders (Ministry of Education)Bio‐X InstitutesShanghai Jiao Tong UniversityShanghai200240China
| |
Collapse
|
11
|
Tahajjodi SS, Yazd EF, Agharahimi A, Akyash F, Hajizadeh-Tafti F, Golzadeh J, Aflatoonian R, Aflatoonian B. Cumulus cells conditioned medium facilitates germ cell differentiation from human embryonic stem cells: An experimental study. Int J Reprod Biomed 2025; 23:33-44. [PMID: 40190455 PMCID: PMC11966215 DOI: 10.18502/ijrm.v23i1.18186] [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: 06/26/2024] [Revised: 10/02/2024] [Accepted: 11/30/2024] [Indexed: 04/09/2025] Open
Abstract
Background Studies have shown thatembryonic stem cells can differentiate into germ cells either spontaneously or directed by the defined factors or by applying a co-culture method or conditioned medium of stromal cells. Objective Here, Yazd4 (human embryonic stem cells [hESC] line; 46, XX) was used to evaluate the effect of human cumulus cells conditioned medium (CCCM) on in vitro germ cell production and development. Materials and Methods In this experimental study 2 different complete media were applied, Dulbecco's modified eagle's medium + 20% fetal bovin serum and embryoid body (EB) medium (KnockOut serum replacement- hESC without basal fibroblast growth factor). Thus, there are 2 control groups for spontaneous differentiation (SD) (SD-EB and SD-DM) and 2 types of CCCM (CCCM-DM and CCCM-EB) as test groups. 40% dilution of the conditioned medium was applied according to previous reports. Results Reverse transcription-quantitative polymerase chain reaction and immunofluorescent staining assays revealed that between groups with similar complete medium, CCCM provides a better condition for in vitro germ cell differentiation from hESCs. Moreover, a comparison between the 2 types of complete media showed that EB medium is more supportive than Dulbecco's modified eagle's medium +20% fetal bovin serum. Conclusion CCCM with EB medium as a complete medium provides a better condition for in vitro germ cell differentiation from hESCs.
Collapse
Affiliation(s)
- Somayyeh Sadat Tahajjodi
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Research and Clinical Center for Infertility, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Reproductive Biology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Ehsan Farashahi Yazd
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Research and Clinical Center for Infertility, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Reproductive Biology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Azam Agharahimi
- Research and Clinical Center for Infertility, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Reproductive Biology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Akyash
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Research and Clinical Center for Infertility, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Reproductive Biology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Hajizadeh-Tafti
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Jalal Golzadeh
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Reza Aflatoonian
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Centre, Royan Institute for Reproductive Biomedicine, Tehran, Iran
| | - Behrouz Aflatoonian
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Research and Clinical Center for Infertility, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Reproductive Biology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
12
|
Aizawa E, Peters AHFM, Wutz A. In vitro gametogenesis: Towards competent oocytes: Limitations and future improvements for generating oocytes from pluripotent stem cells in culture. Bioessays 2025; 47:e2400106. [PMID: 39498732 DOI: 10.1002/bies.202400106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 09/25/2024] [Accepted: 10/10/2024] [Indexed: 11/07/2024]
Abstract
Production of oocytes from pluripotent cell cultures in a dish represents a new paradigm in stem cell and developmental biology and has implications for how we think about life. The spark of life for the next generation occurs at fertilization when sperm and oocyte fuse. In animals, gametes are the only cells that transmit their genomes to the next generation. Oocytes contain in addition a large cytoplasm with factors that direct embryonic development. Reconstitution of mouse oocyte and embryonic development in culture provides experimental opportunities and facilitates an unprecedented understanding of molecular mechanisms. However, the application of in vitro gametogenesis to reproductive medicine or infertility treatment remains challenging. One significant concern is the quality of in vitro-derived oocytes. Here, we review the current understanding and identify limitations in generating oocytes in vitro. From this basis, we explore opportunities for future improvements of the in vitro approach to generating high-quality oocytes.
Collapse
Affiliation(s)
- Eishi Aizawa
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology, ETH Zurich, Zurich, Switzerland
- RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Antoine H F M Peters
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
- Faculty of Sciences, University of Basel, Basel, Switzerland
| | - Anton Wutz
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
13
|
Zhang T, Zhang M, Zhang S, Wang S. Research advances in the construction of stem cell-derived ovarian organoids. Stem Cell Res Ther 2024; 15:505. [PMID: 39736770 DOI: 10.1186/s13287-024-04122-3] [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: 09/19/2024] [Accepted: 12/18/2024] [Indexed: 01/01/2025] Open
Abstract
Ovarian organoids are essential in female reproductive medicine, enhancing our understanding of ovarian diseases and improving treatments, which benefits women's health. Constructing ovarian organoids involves two main processes: differentiating induced pluripotent stem cells (iPSCs) into germ and ovarian somatic cells to restore ovarian function and using extracellular matrix (ECM) to create a suitable ovarian microenvironment and scaffold. Although the technology is still in its early stages, future advancements will likely involve integrating high-throughput analysis, 3D-printed scaffolds, and efficient iPSC induction, driving progress in reproductive and regenerative medicine.
Collapse
Affiliation(s)
- Tianyue Zhang
- Department of Gynecology and Obstetrics, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, People's Republic of China
- Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Mengtong Zhang
- Department of Gynecology and Obstetrics, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, People's Republic of China
| | - Sichen Zhang
- Department of Gynecology and Obstetrics, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, People's Republic of China
| | - Shaowei Wang
- Department of Gynecology and Obstetrics, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, People's Republic of China.
- Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China.
| |
Collapse
|
14
|
Choong E, Dawson EP, Bowman K, Adashi EY. In vitro gametogenesis (IVG): reflections from a workshop. J Assist Reprod Genet 2024; 41:3323-3326. [PMID: 39400648 PMCID: PMC11707097 DOI: 10.1007/s10815-024-03266-8] [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: 07/31/2024] [Accepted: 09/16/2024] [Indexed: 10/15/2024] Open
Abstract
Key ideas from a workshop convened by the National Academies of Sciences, Engineering, and Medicine to discuss developments in IVG (National Academies of Sciences, Engineering, and Medicine 2023) chaired by Dr Eli Y. Adashi (former Dean of Medicine and Biological Sciences at Brown University). The authors are solely responsible for the content of this paper, which does not necessarily represent the views of the National Academies of Sciences, Engineering, and Medicine.
Collapse
Affiliation(s)
| | | | - Katherine Bowman
- National Academies of Sciences, Engineering, and Medicine, Washington, DC, USA
| | | |
Collapse
|
15
|
Li N, Du X, Zhao Y, Zeng Q, Han C, Xiong D, He L, Zhang G, Liu W. Exploring stem cell technology: Pioneering new pathways for female fertility preservation and restoration. Reprod Biol 2024; 24:100958. [PMID: 39393314 DOI: 10.1016/j.repbio.2024.100958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 09/24/2024] [Accepted: 09/28/2024] [Indexed: 10/13/2024]
Abstract
The fertility of women is crucial for the well-being of individuals and families. However, various factors such as chemotherapy, lifestyle changes, among others, may lead to a decline in female fertility, thus emphasizing the significance of preserving and restoring fertility. Stem cells, with their unique capacity for self-renewal and pluripotent differentiation, have made significant strides in areas such as ovarian tissue cryopreservation, in vitro culture of frozen-thawed ovarian tissue, and construction of ovarian-like organs. This review aims to summarize the latest findings in these fields, highlighting the pivotal role, mechanisms, and future prospects of stem cell technology in preserving and restoring female fertility. Additionally, the importance of interdisciplinary collaboration is underscored, as personalized stem cell therapy regimens tailored through interdisciplinary cooperation between reproductive medicine and stem cell fields hold promise in providing reliable solutions for the preservation and restoration of female fertility.
Collapse
Affiliation(s)
- Ningjing Li
- School of Medicine and life sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xinrong Du
- School of Medicine and life sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yuhong Zhao
- College of Laboratory Medicine, Chengdu Medical College, Chengdu 610500, China
| | - Qin Zeng
- Sichuan Provincial Woman's and Children's Hospital / The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu 610045, China
| | - Changli Han
- School of Medicine and life sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Dongsheng Xiong
- Sichuan Provincial Woman's and Children's Hospital / The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu 610045, China
| | - Libing He
- Sichuan Provincial Woman's and Children's Hospital / The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu 610045, China
| | - Guohui Zhang
- Sichuan Provincial Woman's and Children's Hospital / The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu 610045, China.
| | - Weixin Liu
- Sichuan Provincial Woman's and Children's Hospital / The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu 610045, China.
| |
Collapse
|
16
|
Chitnis MS, Gao X, Marlena J, Holle AW. The mechanical journey of primordial germ cells. Am J Physiol Cell Physiol 2024; 327:C1532-C1545. [PMID: 39466178 DOI: 10.1152/ajpcell.00404.2024] [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: 06/13/2024] [Revised: 10/14/2024] [Accepted: 10/14/2024] [Indexed: 10/29/2024]
Abstract
Primordial germ cells (PGCs) are the earliest progenitors of germline cells of the gonads in animals. The tissues that arise from primordial germ cells give rise to male and female gametes and are thus responsible for transmitting genetic information to subsequent generations. The development of gonads, from single cells to fully formed organs, is of great interest to the reproductive biology community. In most higher animals, PGCs are initially specified at a site away from the gonads. They then migrate across multiple tissue microenvironments to reach a mesodermal mass of cells called the genital ridge, where they associate with somatic cells to form sex-specific reproductive organs. Their migratory behavior has been studied extensively to identify which tissues they interact with and how this might affect gonad development. A crucial point overlooked by classical studies has been the physical environment experienced by PGCs as they migrate and the mechanical challenges they might encounter along the way. It has long been understood that migrating cells can sense and adapt to physical forces around them via a variety of mechanisms, and studies have shown that these mechanical signals can guide stem cell fate. In this review, we summarize the mechanical microenvironment of migrating PGCs in different organisms. We describe how cells can adapt to this environment and how this adaptation can influence cell fate. Finally, we propose that mechanical signals play a crucial role in the normal development of the germline and shed light on this unexplored area of developmental biology.
Collapse
Affiliation(s)
- Malhar S Chitnis
- Mechanobiology Institute, National University of Singapore, Singapore
| | - Xu Gao
- Mechanobiology Institute, National University of Singapore, Singapore
- Department of Biomedical Engineering, National University of Singapore, Singapore
| | - Jennifer Marlena
- Mechanobiology Institute, National University of Singapore, Singapore
| | - Andrew W Holle
- Mechanobiology Institute, National University of Singapore, Singapore
- Department of Biomedical Engineering, National University of Singapore, Singapore
| |
Collapse
|
17
|
Naik HC, Chandel D, Majumdar S, Arava M, Baro R, Bv H, Hari K, Ayyamperumal P, Manhas A, Jolly MK, Gayen S. Lineage-specific dynamics of loss of X upregulation during inactive-X reactivation. Stem Cell Reports 2024; 19:1564-1582. [PMID: 39486405 PMCID: PMC11589478 DOI: 10.1016/j.stemcr.2024.10.001] [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: 03/11/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 11/04/2024] Open
Abstract
In mammals, X chromosome dosage is balanced between sexes through the silencing of one X chromosome in females. Recent single-cell RNA sequencing analysis demonstrated that the inactivation of the X chromosome is accompanied by the upregulation of the active X chromosome (Xa) during mouse embryogenesis. Here, we have investigated if the reactivation of inactive-X (Xi) leads to the loss of Xa upregulation in different cellular or developmental contexts. We find that while Xi reactivation and loss of Xa upregulation are tightly coupled in mouse embryonic epiblast and induced pluripotent stem cells, that is not the case in germ cells. Moreover, we demonstrate that partial reactivation of Xi in mouse extra-embryonic endoderm stem cells and human B cells does not result in the loss of Xa upregulation. Finally, we have established a mathematical model for the transcriptional coordination of two X chromosomes. Together, we conclude that the reactivation of Xi is not always synchronized with the loss of Xa upregulation.
Collapse
Affiliation(s)
- Hemant Chandru Naik
- Chromatin, RNA and Genome Laboratory, Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Deepshikha Chandel
- Chromatin, RNA and Genome Laboratory, Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Sudeshna Majumdar
- Chromatin, RNA and Genome Laboratory, Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Maniteja Arava
- Chromatin, RNA and Genome Laboratory, Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Runumi Baro
- Chromatin, RNA and Genome Laboratory, Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Harshavardhan Bv
- Chromatin, RNA and Genome Laboratory, Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore 560012, India; Department of Bioengineering, Indian Institute of Science, Bangalore 560012, India; IISc Mathematics Initiative (IMI), Indian Institute of Science, Bangalore 560012, India
| | - Kishore Hari
- Department of Bioengineering, Indian Institute of Science, Bangalore 560012, India
| | - Parichitran Ayyamperumal
- Chromatin, RNA and Genome Laboratory, Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Avinchal Manhas
- Chromatin, RNA and Genome Laboratory, Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Mohit Kumar Jolly
- Department of Bioengineering, Indian Institute of Science, Bangalore 560012, India
| | - Srimonta Gayen
- Chromatin, RNA and Genome Laboratory, Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore 560012, India.
| |
Collapse
|
18
|
Hutchinson AM, Appeltant R, Burdon T, Bao Q, Bargaje R, Bodnar A, Chambers S, Comizzoli P, Cook L, Endo Y, Harman B, Hayashi K, Hildebrandt T, Korody ML, Lakshmipathy U, Loring JF, Munger C, Ng AHM, Novak B, Onuma M, Ord S, Paris M, Pask AJ, Pelegri F, Pera M, Phelan R, Rosental B, Ryder OA, Sukparangsi W, Sullivan G, Tay NL, Traylor-Knowles N, Walker S, Weberling A, Whitworth DJ, Williams SA, Wojtusik J, Wu J, Ying QL, Zwaka TP, Kohler TN. Advancing stem cell technologies for conservation of wildlife biodiversity. Development 2024; 151:dev203116. [PMID: 39382939 PMCID: PMC11491813 DOI: 10.1242/dev.203116] [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] [Indexed: 10/10/2024]
Abstract
Wildlife biodiversity is essential for healthy, resilient and sustainable ecosystems. For biologists, this diversity also represents a treasure trove of genetic, molecular and developmental mechanisms that deepen our understanding of the origins and rules of life. However, the rapid decline in biodiversity reported recently foreshadows a potentially catastrophic collapse of many important ecosystems and the associated irreversible loss of many forms of life on our planet. Immediate action by conservationists of all stripes is required to avert this disaster. In this Spotlight, we draw together insights and proposals discussed at a recent workshop hosted by Revive & Restore, which gathered experts to discuss how stem cell technologies can support traditional conservation techniques and help protect animal biodiversity. We discuss reprogramming, in vitro gametogenesis, disease modelling and embryo modelling, and we highlight the prospects for leveraging stem cell technologies beyond mammalian species.
Collapse
Affiliation(s)
| | - Ruth Appeltant
- Gamete Research Centre, Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| | - Tom Burdon
- The Roslin Institute, RDSVS, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Qiuye Bao
- IMCB-ESCAR, A*STAR, 61 Biopolis Drive, Proteos, 138673Singapore
| | | | - Andrea Bodnar
- Gloucester Marine Genomics Institute, 417 Main St, Gloucester, MA 01930, USA
| | - Stuart Chambers
- Brightfield Therapeutics, South San Francisco, CA 94080, USA
| | - Pierre Comizzoli
- Smithsonian National Zoo and Conservation Biology Institute, 3001 Connecticut Ave., NW Washington, DC 20008, USA
| | - Laura Cook
- Lawrence Berkeley National Laboratory, 1 Cyclotron Rd, Berkeley, CA 94720, USA
| | - Yoshinori Endo
- University of California San Diego, 9500 Gilman Dr, La Jolla, CA 92093, USA
| | - Bob Harman
- Vet-Stem Inc. & Personalized Stem Cells, Inc., 14261 Danielson Street, Poway, CA 92064, USA
| | | | - Thomas Hildebrandt
- Leibniz Institute for Zoo and Wildlife Research, Alfred-Kowalke-Straße 17, 10315 Berlin, Germany
| | - Marisa L. Korody
- San Diego Zoo Wildlife Alliance, 2920 Zoo Dr, San Diego, CA 92101, USA
| | - Uma Lakshmipathy
- Thermo Fisher Scientific, 168 Third Avenue, Waltham, MA 02451, USA
| | - Jeanne F. Loring
- The Scripps Research Institute, 10550 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Clara Munger
- Department of Biochemistry, University of Cambridge, Hopkins Building, Downing Site, Tennis Court Road, Cambridge CB2 1QW, UK
| | - Alex H. M. Ng
- GC Therapeutics, 610 Main St., North Cambridge, MA 02139, USA
| | - Ben Novak
- Revive & Restore, 1505 Bridgeway, Suite 203, Sausalito, CA 94965, USA
| | - Manabu Onuma
- National Institute for Environmental Studies, 16-2 Onogawa, City of Tsukuba, Ibaraki 305-8506, Japan
| | - Sara Ord
- Colossal Biosciences, 1401 Lavaca St, Unit #155 Austin, TX 78701, USA
| | - Monique Paris
- IBREAM (Institute for Breeding Rare and Endangered African Mammals), Edinburgh EH3 6AT, UK
| | | | - Francisco Pelegri
- University of Wisconsin-Madison, 500 Lincoln Dr, Madison, WI 53706, USA
| | - Martin Pera
- Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Ryan Phelan
- Revive & Restore, 1505 Bridgeway, Suite 203, Sausalito, CA 94965, USA
| | - Benyamin Rosental
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Center for Regenerative Medicine and Stem Cells, Ben Gurion University of the Negev, Beer Sheva 8410501, Israel
| | - Oliver A. Ryder
- San Diego Zoo Wildlife Alliance, 2920 Zoo Dr, San Diego, CA 92101, USA
| | - Woranop Sukparangsi
- Department of Biology, Faculty of Science, Burapha University, 169 Long-Had Bangsaen Rd, Saen Suk, Chon Buri District, Chon Buri 20131, Thailand
| | - Gareth Sullivan
- Department of Pediatric Research, Oslo University Hospital, P.O. Box 4950 Nydalen, N-0424 Oslo, Norway
- School of Medicine, University of St Andrews, North Haugh, St Andrews KY16 9TF, UK
| | | | - Nikki Traylor-Knowles
- Rosenstiel School of Marine, Atmospheric, and Earth Science, University of Miami,4600, Rickenbacker Cswy, Key Biscayne, FL 33149, USA
| | - Shawn Walker
- ViaGen Pets & Equine, PO Box 1119, Cedar Park, TX 78613, USA
| | | | - Deanne J. Whitworth
- University of Queensland, Sir Fred Schonell Drive, Brisbane, Queensland, 4072, Australia
| | | | - Jessye Wojtusik
- Omaha's Henry Doorly Zoo & Aquarium, 3701 S 10th St, Omaha, NE 68107, USA
| | - Jun Wu
- University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Qi-Long Ying
- Keck School of Medicine of University of Southern California, 1975 Zonal Ave, Los Angeles, CA 90033, USA
| | - Thomas P. Zwaka
- Department of Cell, Developmental, and Regenerative Biology, and Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Timo N. Kohler
- Department of Biochemistry, University of Cambridge, Hopkins Building, Downing Site, Tennis Court Road, Cambridge CB2 1QW, UK
| |
Collapse
|
19
|
Shimada R, Ishiguro K. Female-specific mechanisms of meiotic initiation and progression in mammalian oocyte development. Genes Cells 2024; 29:797-807. [PMID: 39119753 PMCID: PMC11555627 DOI: 10.1111/gtc.13152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 06/16/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024]
Abstract
Meiosis is regulated in sexually dimorphic manners in mammals. In females, the commitment to and entry into meiosis are coordinated with the developmental program of oocytes. Female germ cells initiate meiosis within a short time window during the fetal period and then undergo meiotic arrest until puberty. However, the genetic mechanisms underlying the orchestration of oocyte development and meiosis to maximize the reproductive lifespan of mammalian females remain largely elusive. While meiotic initiation is regulated by a sexually common mechanism, where meiosis initiator and Stimulated by Retinoic Acid Gene 8 (STRA8) activate the meiotic genes, the female-specific mode of meiotic initiation is mediated by the interaction between retinoblastoma (RB) and STRA8. This review highlights the female-specific mechanisms of meiotic initiation and meiotic prophase progression in the context of oocyte development. Furthermore, the downstream pathway of the RB-STRA8 interaction that may regulate meiotic arrest will be discussed in the context of oocyte development, highlighting a potential genetic link between the female-specific mode of meiotic entry and meiotic arrest.
Collapse
Affiliation(s)
- Ryuki Shimada
- Department of Chromosome Biology, Institute of Molecular Embryology and Genetics (IMEG)Kumamoto UniversityKumamotoJapan
| | - Kei‐ichiro Ishiguro
- Department of Chromosome Biology, Institute of Molecular Embryology and Genetics (IMEG)Kumamoto UniversityKumamotoJapan
| |
Collapse
|
20
|
Aquino LVCD, Olindo SL, Silva YLFE, Oliveira LRMD, Moura YBF, Rodrigues ALR, Praxedes ÉA, Oliveira MFD, Silva AR, Pereira AF. Cryopreservation and passaging optimization for Galea spixii (Wagler, 1831) adult skin fibroblast lines: A step forward in species management and genetic studies. Acta Histochem 2024; 126:152185. [PMID: 39059228 DOI: 10.1016/j.acthis.2024.152185] [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: 03/22/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND In vitro culture of fibroblasts is a technique based on cell isolation, physiological characterization, and cryopreservation. This technique has not been described for Galea spixii, therefore, it can be used to learn about its cellular biology and genetic diversity. OBJECTIVE We established fibroblast lines of six G. spixii individuals from several passages (second, fifth, eighth, and tenth) and cryopreserved them. METHODS Fibroblasts recovered from skin biopsies were identified based on morphology, immunocytochemistry, and karyotyping. The cells were analyzed for morphology, ultrastructure, viability, proliferation, metabolism, oxidative stress, bioenergetic potential, and apoptosis before and after cryopreservation. RESULTS After the eighth passage, the fibroblasts showed morphological and karyotypic changes, although their viability, metabolism, and proliferation did not change. An increase in oxidative stress and bioenergetic potential from the fifth to the eighth passages were also observed. Post cryopreservation, cell damage with respect to the ultrastructure, viability, proliferative rate, apoptotic levels, oxidative stress, and bioenergetic potential were verified. CONCLUSION Fibroblasts up to the tenth passage could be cultured in vitro. However, cells at the fifth passage were of better quality to be used for reproductive techniques. Additionally, optimization of the cryopreservation protocol is essential to improve the physiological parameters of these cells.
Collapse
Affiliation(s)
| | - Samara Lima Olindo
- Laboratory of Animal Biotechnology, Federal Rural University of Semi-Arid, Mossoró, RN, Brazil.
| | | | | | | | | | - Érika Almeida Praxedes
- Laboratory of Animal Biotechnology, Federal Rural University of Semi-Arid, Mossoró, RN, Brazil. erikaalmeida-@hotmail.com
| | - Moacir Franco de Oliveira
- Laboratory of Applied Animal Morphophysiology, Federal Rural University of Semi-Arid, Mossoró, RN, Brazil.
| | - Alexandre Rodrigues Silva
- Laboratory of Animal Germplasma Conservation, Federal Rural University of Semi-Arid, Mossoró, RN, Brazil.
| | | |
Collapse
|
21
|
Villalba A. Artificial Gametes and Human Reproduction in the 21st Century: An Ethical Analysis. Reprod Sci 2024; 31:2174-2183. [PMID: 38780744 DOI: 10.1007/s43032-024-01558-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 04/11/2024] [Indexed: 05/25/2024]
Abstract
Artificial gametes, derived from stem cells, have the potential to enable in vitro fertilization of embryos. Currently, artificial gametes are only being generated in laboratory animals; however, considerable efforts are underway to develop artificial gametes using human cell sources. These artificial gametes are being proposed as a means to address infertility through assisted reproductive technologies. Nonetheless, the availability of artificial gametes obtained from adult organisms can potentially expand the possibilities of reproduction. Various groups, such as same-sex couples, post-menopausal women, and deceased donors, could potentially utilize artificial gametes to conceive genetically related offspring. The advent of artificial gametes raises significant bioethical questions. Should all these reproductive scenarios be accepted? How can we delineate the range of future reproductive choices? A normative bioethical framework may be necessary to establish a consensus regarding the use of human artificial gametes. This review aims to present the current state of research on the biological roadmap for generating artificial gametes, while also summarizing proposed approaches to establish a normative framework that delineates ethically acceptable paths for reproduction.
Collapse
Affiliation(s)
- Adrian Villalba
- Departamento de Filosofía II, Universidad de Granada, Granada, Spain.
| |
Collapse
|
22
|
Jang SW, Kim YR, Han JH, Jang H, Choi HW. Generation of mouse and rat xenogeneic ovaries in vitro for production of mouse oocyte. Anim Cells Syst (Seoul) 2024; 28:303-314. [PMID: 38868077 PMCID: PMC11168328 DOI: 10.1080/19768354.2024.2363601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 05/30/2024] [Indexed: 06/14/2024] Open
Abstract
The system forming ovarian follicles is developed to investigate in vitro folliculogenesis in a confined environment to obtain functional oocytes. Several studies have reported the successful generation of fully functional oocytes using mouse-induced pluripotent stem cells (iPSCs) and mouse female germline stem cells (fGSCs) as sources of stem cells for in vitro gametogenesis models. In addition, human oogonia have been generated through heterologous co-culture of differentiated human primordial germ cell-like cells (hPGCLCs) with mouse germline somatic cells, although oocyte formation remains challenging. Thus, studies on in vitro ovarian formation in other species are utilized as an introductory approach for in vitro mammalian gametogenesis by understanding the differences in culture systems between species and underlying mechanisms. In this study, we optimized the method of the entire oogenesis process from rat embryonic gonads. We identified well-maturated MII oocytes from rat gonads using our constructed method. Moreover, we generated the first successful in vitro reconstitution of xenogeneic follicles from mouse primordial germ cells (PGCs) and rat somatic cells. We also established an appropriate culture medium and incubation period for xenogeneic follicles. This method will be helpful in studies of xenogeneic follicular development and oocyte generation.
Collapse
Affiliation(s)
- Si Won Jang
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju, Republic of Korea
| | - Ye Rim Kim
- Department of Animal Science, Jeonbuk National University, Jeonju, Republic of Korea
| | - Jae Ho Han
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju, Republic of Korea
| | - Hoon Jang
- Department of Life Science, Jeonbuk National University, Jeonju, Republic of Korea
| | - Hyun Woo Choi
- Department of Agricultural Convergence Technology, Jeonbuk National University, Jeonju, Republic of Korea
- Department of Animal Science, Jeonbuk National University, Jeonju, Republic of Korea
| |
Collapse
|
23
|
Kurlovich J, Rodriguez Polo I, Dovgusha O, Tereshchenko Y, Cruz CRV, Behr R, Günesdogan U. Generation of marmoset primordial germ cell-like cells under chemically defined conditions. Life Sci Alliance 2024; 7:e202302371. [PMID: 38499329 PMCID: PMC10948935 DOI: 10.26508/lsa.202302371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 03/20/2024] Open
Abstract
Primordial germ cells (PGCs) are the embryonic precursors of sperm and oocytes, which transmit genetic/epigenetic information across generations. Mouse PGC and subsequent gamete development can be fully reconstituted in vitro, opening up new avenues for germ cell studies in biomedical research. However, PGCs show molecular differences between rodents and humans. Therefore, to establish an in vitro system that is closely related to humans, we studied PGC development in vivo and in vitro in the common marmoset monkey Callithrix jacchus (cj). Gonadal cjPGCs at embryonic day 74 express SOX17, AP2Ɣ, BLIMP1, NANOG, and OCT4A, which is reminiscent of human PGCs. We established transgene-free induced pluripotent stem cell (cjiPSC) lines from foetal and postnatal fibroblasts. These cjiPSCs, cultured in defined and feeder-free conditions, can be differentiated into precursors of mesendoderm and subsequently into cjPGC-like cells (cjPGCLCs) with a transcriptome similar to human PGCs/PGCLCs. Our results not only pave the way for studying PGC development in a non-human primate in vitro under experimentally controlled conditions, but also provide the opportunity to derive functional marmoset gametes in future studies.
Collapse
Affiliation(s)
- Julia Kurlovich
- Göttingen Center for Molecular Biosciences, Department of Developmental Biology, University of Göttingen, Göttingen, Germany
| | - Ignacio Rodriguez Polo
- Göttingen Center for Molecular Biosciences, Department of Developmental Biology, University of Göttingen, Göttingen, Germany
- German Primate Center-Leibniz Institute for Primate Research, Research Platform Degenerative Diseases, Göttingen, Germany
- Stem Cell and Human Development Laboratory, The Francis Crick Institute, London, UK
| | - Oleksandr Dovgusha
- Göttingen Center for Molecular Biosciences, Department of Developmental Biology, University of Göttingen, Göttingen, Germany
| | - Yuliia Tereshchenko
- German Primate Center-Leibniz Institute for Primate Research, Research Platform Degenerative Diseases, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
| | - Carmela Rieline V Cruz
- Göttingen Center for Molecular Biosciences, Department of Developmental Biology, University of Göttingen, Göttingen, Germany
| | - Rüdiger Behr
- German Primate Center-Leibniz Institute for Primate Research, Research Platform Degenerative Diseases, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
| | - Ufuk Günesdogan
- Göttingen Center for Molecular Biosciences, Department of Developmental Biology, University of Göttingen, Göttingen, Germany
- Department for Molecular Developmental Biology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| |
Collapse
|
24
|
Pasquariello R, Bogliolo L, Di Filippo F, Leoni GG, Nieddu S, Podda A, Brevini TAL, Gandolfi F. Use of assisted reproductive technologies (ARTs) to shorten the generational interval in ruminants: current status and perspectives. Theriogenology 2024; 225:16-32. [PMID: 38788626 DOI: 10.1016/j.theriogenology.2024.05.026] [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: 03/05/2024] [Revised: 05/18/2024] [Accepted: 05/18/2024] [Indexed: 05/26/2024]
Abstract
The challenges posed by climate change and increasing world population are stimulating renewed efforts for improving the sustainability of animal production. To meet such challenges, the contribution of genomic selection approaches, in combination with assisted reproductive technologies (ARTs), to spreading and preserving animal genetics is essential. The largest increase in genetic gain can be achieved by shortening the generation interval. This review provides an overview of the current status and progress of advanced ARTs that could be applied to reduce the generation time in both female and male of domestic ruminants. In females, the use of juvenile in vitro embryo transfer (JIVET) enables to generate offspring after the transfer of in vitro produced embryos derived from oocytes of prepubertal genetically superior donors reducing the generational interval and acceleration genetic gain. The current challenge is increasing in vitro embryo production (IVEP) from prepubertal derived oocytes which is still low and variable. The two main factors limiting IVEP success are the intrinsic quality of prepubertal oocytes and the culture systems for in vitro maturation (IVM). In males, advancements in ARTs are providing new strategies to in vitro propagate spermatogonia and differentiate them into mature sperm or even to recapitulate the whole process of spermatogenesis from embryonic stem cells. Moreover, the successful use of immature cells, such as round spermatids, for intracytoplasmic injection (ROSI) and IVEP could allow to complete the entire process in few months. However, these approaches have been successfully applied to human and mouse whereas only a few studies have been published in ruminants and results are still controversial. This is also dependent on the efficiency of ROSI that is limited by the current isolation and selection protocols of round spermatids. In conclusion, the current efforts for improving these reproductive methodologies could lead toward a significant reduction of the generational interval in livestock animals that could have a considerable impact on agriculture sustainability.
Collapse
Affiliation(s)
- Rolando Pasquariello
- Department of Agricultural and Environmental Sciences, University of Milan, Milano, Italy
| | - Luisa Bogliolo
- Department of Veterinary Medicine, University of Sassari, Sassari, Italy
| | - Francesca Di Filippo
- Department of Agricultural and Environmental Sciences, University of Milan, Milano, Italy
| | | | - Stefano Nieddu
- Department of Veterinary Medicine, University of Sassari, Sassari, Italy
| | - Andrea Podda
- Department of Veterinary Medicine, University of Sassari, Sassari, Italy
| | - Tiziana A L Brevini
- Laboratory of Biomedical Embryology and Tissue Engineering, Department of Veterinary Medicine and Animal Science, University of Milan, Lodi, Italy
| | - Fulvio Gandolfi
- Department of Agricultural and Environmental Sciences, University of Milan, Milano, Italy.
| |
Collapse
|
25
|
de Castro RCF, Buranello TW, Recchia K, de Souza AF, Pieri NCG, Bressan FF. Emerging Contributions of Pluripotent Stem Cells to Reproductive Technologies in Veterinary Medicine. J Dev Biol 2024; 12:14. [PMID: 38804434 PMCID: PMC11130827 DOI: 10.3390/jdb12020014] [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: 01/28/2024] [Revised: 04/10/2024] [Accepted: 04/22/2024] [Indexed: 05/29/2024] Open
Abstract
The generation of mature gametes and competent embryos in vitro from pluripotent stem cells has been successfully achieved in a few species, mainly in mice, with recent advances in humans and scarce preliminary reports in other domestic species. These biotechnologies are very attractive as they facilitate the understanding of developmental mechanisms and stages that are generally inaccessible during early embryogenesis, thus enabling advanced reproductive technologies and contributing to the generation of animals of high genetic merit in a short period. Studies on the production of in vitro embryos in pigs and cattle are currently used as study models for humans since they present more similar characteristics when compared to rodents in both the initial embryo development and adult life. This review discusses the most relevant biotechnologies used in veterinary medicine, focusing on the generation of germ-cell-like cells in vitro through the acquisition of totipotent status and the production of embryos in vitro from pluripotent stem cells, thus highlighting the main uses of pluripotent stem cells in livestock species and reproductive medicine.
Collapse
Affiliation(s)
- Raiane Cristina Fratini de Castro
- Department of Surgery, Faculty of Veterinary Medicine and Animal Sciences, University of Sao Paulo, São Paulo 01001-010, SP, Brazil; (R.C.F.d.C.); (T.W.B.); (K.R.)
| | - Tiago William Buranello
- Department of Surgery, Faculty of Veterinary Medicine and Animal Sciences, University of Sao Paulo, São Paulo 01001-010, SP, Brazil; (R.C.F.d.C.); (T.W.B.); (K.R.)
| | - Kaiana Recchia
- Department of Surgery, Faculty of Veterinary Medicine and Animal Sciences, University of Sao Paulo, São Paulo 01001-010, SP, Brazil; (R.C.F.d.C.); (T.W.B.); (K.R.)
| | - Aline Fernanda de Souza
- Department of Veterinary Medicine, School of Animal Sciences and Food Engineering, University of Sao Paulo, Pirassununga 13635-900, SP, Brazil;
| | - Naira Caroline Godoy Pieri
- Department of Veterinary Medicine, School of Animal Sciences and Food Engineering, University of Sao Paulo, Pirassununga 13635-900, SP, Brazil;
| | - Fabiana Fernandes Bressan
- Department of Surgery, Faculty of Veterinary Medicine and Animal Sciences, University of Sao Paulo, São Paulo 01001-010, SP, Brazil; (R.C.F.d.C.); (T.W.B.); (K.R.)
- Department of Veterinary Medicine, School of Animal Sciences and Food Engineering, University of Sao Paulo, Pirassununga 13635-900, SP, Brazil;
| |
Collapse
|
26
|
Silber SJ, Goldsmith S, Castleman L, Hayashi K. In Vitro Maturation, In Vitro Oogenesis, and Ovarian Longevity. Reprod Sci 2024; 31:1234-1245. [PMID: 38160209 PMCID: PMC11090930 DOI: 10.1007/s43032-023-01427-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 12/11/2023] [Indexed: 01/03/2024]
Abstract
This paper will review a remarkable new approach to in vitro maturation "IVM" of oocytes from ovarian tissue, based on our results with in vitro oogenesis from somatic cells. As an aside benefit we also have derived a better understanding of ovarian longevity from ovary transplant. We have found that primordial follicle recruitment is triggered by tissue pressure gradients. Increased pressure holds the follicle in meiotic arrest and prevents recruitment. Therefore recruitment occurs first in the least dense inner tissue of the cortico-medullary junction. Many oocytes can be obtained from human ovarian tissue and mature to metaphase 2 in vitro with no need for ovarian stimulation. Ovarian stimulation may only be necessary for removing the oocyte from the ovary, but this can also be accomplished by simple dissection at the time of ovary tissue cryopreservation. By using surgical dissection of the removed ovary, rather than a needle stick, we can obtain many oocytes from very small follicles not visible with ultrasound. A clearer understanding of ovarian function has come from in vitro oogenesis experiments, and that explains why IVM has now become so simple and robust. Tissue pressure (and just a few "core genes" in the mouse) direct primordial follicle recruitment and development to mature oocyte, and therefore also control ovarian longevity. There are three distinct phases to oocyte development both in vitro and in vivo: in vitro differentiation "IVD" which is not gonadotropin sensitive (the longest phase), in vitro gonadotropin sensitivity "IVG" which is the phase of gonadotropin stimulation to prepare for meiotic competence, and IVM to metaphase II. On any given day 35% of GVs in ovarian tissue have already undergone "IVD" and "IVG" in vivo, and therefore are ready for IVM.
Collapse
Affiliation(s)
- Sherman J Silber
- Infertility Center of St. Louis at St. Luke's Hospital, St. Louis, MO, 63017, USA.
| | - Sierra Goldsmith
- Infertility Center of St. Louis at St. Luke's Hospital, St. Louis, MO, 63017, USA.
| | - Leilani Castleman
- Infertility Center of St. Louis at St. Luke's Hospital, St. Louis, MO, 63017, USA
| | - Katsuhiko Hayashi
- Department of Genome Biology, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
| |
Collapse
|
27
|
Barton LJ, Roa-de la Cruz L, Lehmann R, Lin B. The journey of a generation: advances and promises in the study of primordial germ cell migration. Development 2024; 151:dev201102. [PMID: 38607588 PMCID: PMC11165723 DOI: 10.1242/dev.201102] [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] [Indexed: 04/13/2024]
Abstract
The germline provides the genetic and non-genetic information that passes from one generation to the next. Given this important role in species propagation, egg and sperm precursors, called primordial germ cells (PGCs), are one of the first cell types specified during embryogenesis. In fact, PGCs form well before the bipotential somatic gonad is specified. This common feature of germline development necessitates that PGCs migrate through many tissues to reach the somatic gonad. During their journey, PGCs must respond to select environmental cues while ignoring others in a dynamically developing embryo. The complex multi-tissue, combinatorial nature of PGC migration is an excellent model for understanding how cells navigate complex environments in vivo. Here, we discuss recent findings on the migratory path, the somatic cells that shepherd PGCs, the guidance cues somatic cells provide, and the PGC response to these cues to reach the gonad and establish the germline pool for future generations. We end by discussing the fate of wayward PGCs that fail to reach the gonad in diverse species. Collectively, this field is poised to yield important insights into emerging reproductive technologies.
Collapse
Affiliation(s)
- Lacy J. Barton
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249, USA
| | - Lorena Roa-de la Cruz
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, One UTSA Circle, San Antonio, TX 78249, USA
| | - Ruth Lehmann
- Whitehead Institute and Department of Biology, MIT, 455 Main Street, Cambridge, MA 02142, USA
| | - Benjamin Lin
- Department of Biochemistry & Cell Biology, Stony Brook University, Stony Brook, NY, 11794, USA
| |
Collapse
|
28
|
Sepulveda-Rincon LP, Wang YF, Whilding C, Moyon B, Ojarikre OA, Maciulyte V, Hamazaki N, Hayashi K, Turner JMA, Leitch HG. Determining the potency of primordial germ cells by injection into early mouse embryos. Dev Cell 2024; 59:695-704.e5. [PMID: 38359835 DOI: 10.1016/j.devcel.2024.01.022] [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: 11/09/2022] [Revised: 11/29/2023] [Accepted: 01/24/2024] [Indexed: 02/17/2024]
Abstract
Primordial germ cells (PGCs) are the earliest precursors of the gametes. During normal development, PGCs only give rise to oocytes or spermatozoa. However, PGCs can acquire pluripotency in vitro by forming embryonic germ (EG) cells and in vivo during teratocarcinogenesis. Classic embryological experiments directly assessed the potency of PGCs by injection into the pre-implantation embryo. As no contribution to embryos or adult mice was observed, PGCs have been described as unipotent. Here, we demonstrate that PGCs injected into 8-cell embryos can initially survive, divide, and contribute to the developing inner cell mass. Apoptosis-deficient PGCs exhibit improved survival in isolated epiblasts and can form naive pluripotent embryonic stem cell lines. However, contribution to the post-implantation embryo is limited, with no functional incorporation observed. In contrast, PGC-like cells show an extensive contribution to mid-gestation chimeras. We thus propose that PGC formation in vivo establishes a latent form of pluripotency that restricts chimera contribution.
Collapse
Affiliation(s)
- Lessly P Sepulveda-Rincon
- Medical Research Council (MRC) Laboratory of Medical Sciences (LMS), London W12 0HS, UK; Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London W12 0HS, UK.
| | - Yi-Fang Wang
- Medical Research Council (MRC) Laboratory of Medical Sciences (LMS), London W12 0HS, UK
| | - Chad Whilding
- Medical Research Council (MRC) Laboratory of Medical Sciences (LMS), London W12 0HS, UK
| | - Benjamin Moyon
- Medical Research Council (MRC) Laboratory of Medical Sciences (LMS), London W12 0HS, UK
| | - Obah A Ojarikre
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Valdone Maciulyte
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Nobuhiko Hamazaki
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka 812-8582, Japan
| | - Katsuhiko Hayashi
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka 812-8582, Japan; Department of Genome Biology, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita 565-0871, Japan; Graduate School of Frontier Biosciences, Osaka University, Yamadaoka 2-2, Suita 565-0871, Japan; Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Yamadaoka 2-2, Suita 565-0871, Japan
| | - James M A Turner
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Harry G Leitch
- Medical Research Council (MRC) Laboratory of Medical Sciences (LMS), London W12 0HS, UK; Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London W12 0HS, UK.
| |
Collapse
|
29
|
Rodriguez-Polo I, Moris N. Using Embryo Models to Understand the Development and Progression of Embryonic Lineages: A Focus on Primordial Germ Cell Development. Cells Tissues Organs 2024; 213:503-522. [PMID: 38479364 PMCID: PMC7616515 DOI: 10.1159/000538275] [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: 11/30/2023] [Accepted: 03/05/2024] [Indexed: 05/03/2024] Open
Abstract
BACKGROUND Recapitulating mammalian cell type differentiation in vitro promises to improve our understanding of how these processes happen in vivo, while bringing additional prospects for biomedical applications. The establishment of stem cell-derived embryo models and embryonic organoids, which have experienced explosive growth over the last few years, opens new avenues for research due to their scale, reproducibility, and accessibility. Embryo models mimic various developmental stages, exhibit different degrees of complexity, and can be established across species. Since embryo models exhibit multiple lineages organized spatially and temporally, they are likely to provide cellular niches that, to some degree, recapitulate the embryonic setting and enable "co-development" between cell types and neighbouring populations. One example where this is already apparent is in the case of primordial germ cell-like cells (PGCLCs). SUMMARY While directed differentiation protocols enable the efficient generation of high PGCLC numbers, embryo models provide an attractive alternative as they enable the study of interactions of PGCLCs with neighbouring cells, alongside the regulatory molecular and biophysical mechanisms of PGC competency. Additionally, some embryo models can recapitulate post-specification stages of PGC development (including migration or gametogenesis), mimicking the inductive signals pushing PGCLCs to mature and differentiate and enabling the study of PGCLC development across stages. Therefore, in vitro models may allow us to address questions of cell type differentiation, and PGC development specifically, that have hitherto been out of reach with existing systems. KEY MESSAGE This review evaluates the current advances in stem cell-based embryo models, with a focus on their potential to model cell type-specific differentiation in general and in particular to address open questions in PGC development and gametogenesis.
Collapse
Affiliation(s)
| | - Naomi Moris
- The Francis Crick Institute, 1 Midland Road, Somers Town, London, NW1 1AT, UK
| |
Collapse
|
30
|
Cowl VB, Comizzoli P, Appeltant R, Bolton RL, Browne RK, Holt WV, Penfold LM, Swegen A, Walker SL, Williams SA. Cloning for the Twenty-First Century and Its Place in Endangered Species Conservation. Annu Rev Anim Biosci 2024; 12:91-112. [PMID: 37988633 DOI: 10.1146/annurev-animal-071423-093523] [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] [Indexed: 11/23/2023]
Abstract
Cloning as it relates to the animal kingdom generally refers to the production of genetically identical individuals. Because cloning is increasingly the subject of renewed attention as a tool for rescuing endangered or extinct species, it seems timely to dissect the role of the numerous reproductive techniques encompassed by this term in animal species conservation. Although cloning is typically associated with somatic cell nuclear transfer, the recent advent of additional techniques that allow genome replication without genetic recombination demands that the use of induced pluripotent stem cells to generate gametes or embryos, as well as older methods such as embryo splitting, all be included in this discussion. Additionally, the phenomenon of natural cloning (e.g., a subset of fish, birds, invertebrates, and reptilian species that reproduce via parthenogenesis) must also be pointed out. Beyond the biology of these techniques are practical considerations and the ethics of using cloning and associated procedures in endangered or extinct species. All of these must be examined in concert to determine whether cloning has a place in species conservation. Therefore, we synthesize progress in cloning and associated techniques and dissect the practical and ethical aspects of these methods as they pertain to endangered species conservation.
Collapse
Affiliation(s)
- Veronica B Cowl
- North of England Zoological Society (Chester Zoo), Chester, United Kingdom;
- European Association of Zoos and Aquaria, Amsterdam, The Netherlands
| | - Pierre Comizzoli
- Smithsonian's National Zoo and Conservation Biology Institute, Washington, DC, USA;
| | - Ruth Appeltant
- Gamete Research Centre, Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium;
| | | | - Robert K Browne
- Sustainability America, Sarteneja, Corozal District, Belize;
| | - William V Holt
- Department of Oncology and Metabolism, The Medical School, University of Sheffield, Sheffield, United Kingdom;
| | - Linda M Penfold
- South East Zoo Alliance for Reproduction & Conservation, Yulee, Florida, USA;
| | - Aleona Swegen
- Priority Research Centre for Reproductive Science, University of Newcastle, Callaghan, New South Wales, Australia;
| | - Susan L Walker
- North of England Zoological Society (Chester Zoo), Chester, United Kingdom;
- Nature's SAFE, Whitchurch, Shropshire, United Kingdom;
| | - Suzannah A Williams
- Nature's SAFE, Whitchurch, Shropshire, United Kingdom;
- Nuffield Department of Women's and Reproductive Health, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom;
| |
Collapse
|
31
|
Frost ER, Gilchrist RB. Making human eggs in a dish: are we close? Trends Biotechnol 2024; 42:168-178. [PMID: 37625913 DOI: 10.1016/j.tibtech.2023.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/05/2023] [Accepted: 07/30/2023] [Indexed: 08/27/2023]
Abstract
In the space of 50 years, we have seen incredible achievements in human reproductive medicine. With these leaps forward, it is no wonder that there is a major interest in women's reproductive health research, including extension of reproductive lifespan. Substantial effort is currently being made to address this challenge, including from the commercial sector. In vitro gametogenesis (IVG) in mice is a spectacular breakthrough and has the potential to offer hope to women with intractable infertility. However, with such lofty goals, some reflection may be called for: mastering all of the techniques required for complete and safe IVG in women is likely to be extraordinarily difficult.
Collapse
Affiliation(s)
- Emily R Frost
- Fertility & Research Centre, Discipline of Women's Health, School of Clinical Medicine, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Robert B Gilchrist
- Fertility & Research Centre, Discipline of Women's Health, School of Clinical Medicine, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
32
|
Bao Q, Tay NL, Lim CY, Chua DHH, Kee SK, Choolani M, Loh YH, Ng SC, Chai C. Integration-free induced pluripotent stem cells from three endangered Southeast Asian non-human primate species. Sci Rep 2024; 14:2391. [PMID: 38287040 PMCID: PMC10825216 DOI: 10.1038/s41598-023-50510-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 12/20/2023] [Indexed: 01/31/2024] Open
Abstract
Advanced molecular and cellular technologies provide promising tools for wildlife and biodiversity conservation. Induced pluripotent stem cell (iPSC) technology offers an easily accessible and infinite source of pluripotent stem cells, and have been derived from many threatened wildlife species. This paper describes the first successful integration-free reprogramming of adult somatic cells to iPSCs, and their differentiation, from three endangered Southeast Asian primates: the Celebes Crested Macaque (Macaca nigra), the Lar Gibbon (Hylobates lar), and the Siamang (Symphalangus syndactylus). iPSCs were also generated from the Proboscis Monkey (Nasalis larvatus). Differences in mechanisms could elicit new discoveries regarding primate evolution and development. iPSCs from endangered species provides a safety net in conservation efforts and allows for sustainable sampling for research and conservation, all while providing a platform for the development of further in vitro models of disease.
Collapse
Affiliation(s)
- Qiuye Bao
- Institute of Molecular and Cell Biology-Endangered Species Conservation By Assisted Reproduction (IMCB-ESCAR) Joint Laboratory, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Singapore, 138673, Singapore
| | - Nicole Liling Tay
- Institute of Molecular and Cell Biology-Endangered Species Conservation By Assisted Reproduction (IMCB-ESCAR) Joint Laboratory, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Singapore, 138673, Singapore
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
| | - Christina Yingyan Lim
- Institute of Molecular and Cell Biology-Endangered Species Conservation By Assisted Reproduction (IMCB-ESCAR) Joint Laboratory, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Singapore, 138673, Singapore
| | | | - Su Keyau Kee
- Cytogenetics Laboratory, Department of Pathology, Singapore General Hospital, 20 College Road, Singapore, 169856, Singapore
| | - Mahesh Choolani
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
| | - Yuin-Han Loh
- Institute of Molecular and Cell Biology-Endangered Species Conservation By Assisted Reproduction (IMCB-ESCAR) Joint Laboratory, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Singapore, 138673, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, 117543, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117593, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, 28 Medical Drive, Singapore, 117456, Singapore
| | - Soon Chye Ng
- Institute of Molecular and Cell Biology-Endangered Species Conservation By Assisted Reproduction (IMCB-ESCAR) Joint Laboratory, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Singapore, 138673, Singapore.
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore.
- Sincere Healthcare Group, 8 Sinaran Drive, Singapore, 307470, Singapore.
| | - Chou Chai
- Institute of Molecular and Cell Biology-Endangered Species Conservation By Assisted Reproduction (IMCB-ESCAR) Joint Laboratory, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Singapore, 138673, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore, 308232, Singapore
| |
Collapse
|
33
|
Nogueira IPM, Costa GMJ, Lacerda SMDSN. Avian iPSC Derivation to Recover Threatened Wild Species: A Comprehensive Review in Light of Well-Established Protocols. Animals (Basel) 2024; 14:220. [PMID: 38254390 PMCID: PMC10812705 DOI: 10.3390/ani14020220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/24/2024] Open
Abstract
Induced pluripotent stem cells (iPSCs) were first generated by Yamanaka in 2006, revolutionizing research by overcoming limitations imposed by the use of embryonic stem cells. In terms of the conservation of endangered species, iPSC technology presents itself as a viable alternative for the manipulation of target genetics without compromising specimens. Although iPSCs have been successfully generated for various species, their application in nonmammalian species, particularly avian species, requires further in-depth investigation to cover the diversity of wild species at risk and their different protocol requirements. This study aims to provide an overview of the workflow for iPSC induction, comparing well-established protocols in humans and mice with the limited information available for avian species. Here, we discuss the somatic cell sources to be reprogrammed, genetic factors, delivery methods, enhancers, a brief history of achievements in avian iPSC derivation, the main approaches for iPSC characterization, and the future perspectives and challenges for the field. By examining the current protocols and state-of-the-art techniques employed in iPSC generation, we seek to contribute to the development of efficient and species-specific iPSC methodologies for at-risk avian species. The advancement of iPSC technology holds great promise for achieving in vitro germline competency and, consequently, addressing reproductive challenges in endangered species, providing valuable tools for basic research, bird genetic preservation and rescue, and the establishment of cryobanks for future conservation efforts.
Collapse
Affiliation(s)
| | | | - Samyra Maria dos Santos Nassif Lacerda
- Laboratory of Cellular Biology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (I.P.M.N.); (G.M.J.C.)
| |
Collapse
|
34
|
Oikawa M, Hirabayashi M, Kobayashi T. Induction of Primordial Germ Cell-Like Cells from Rat Pluripotent Stem Cells. Methods Mol Biol 2024; 2770:99-111. [PMID: 38351449 DOI: 10.1007/978-1-0716-3698-5_8] [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] [Indexed: 02/16/2024]
Abstract
In vitro induction of primordial germ cell like-cells (PGCLCs) from pluripotent stem cells (PSCs) is a robust method that will contribute to understanding the fundamentals of cell fate decisions, animal breeding, and future reproductive medicine. Here, we introduce this system established in the rat model. We describe a stepwise protocol to induce epiblast-like cells and subsequent PGCLCs by forming spherical aggregates from rat PSCs. We also describe a protocol to mature these PGCLCs from specified/migratory to the gonadal stage by aggregation with female gonadal somatic cells.
Collapse
Affiliation(s)
- Mami Oikawa
- Division of Mammalian Embryology, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Laboratory of Regenerative Medicine, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Masumi Hirabayashi
- Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Aichi, Japan
- The Graduate University of Advanced Studies, Aichi, Japan
| | - Toshihiro Kobayashi
- Division of Mammalian Embryology, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
- Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Aichi, Japan.
| |
Collapse
|
35
|
Kanegi R, Hatoya S, Kimura K, Yodoe K, Nishimura T, Sugiura K, Kawate N, Inaba T. Generation, characterization, and differentiation of induced pluripotent stem-like cells in the domestic cat. J Reprod Dev 2023; 69:317-327. [PMID: 37880086 PMCID: PMC10721851 DOI: 10.1262/jrd.2022-038] [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: 03/23/2022] [Accepted: 09/28/2023] [Indexed: 10/27/2023] Open
Abstract
Induced pluripotent stem (iPS) cells are generated from somatic cells and can differentiate into various cell types. Therefore, these cells are expected to be a powerful tool for modeling diseases and transplantation therapy. Generation of domestic cat iPS cells depending on leukemia inhibitory factor has been reported; however, this strategy may not be optimized. Considering that domestic cats are excellent models for studying spontaneous diseases, iPS cell generation is crucial. In this study, we aimed to derive iPS cells from cat embryonic fibroblasts retrovirally transfected with mouse Oct3/4, Klf4, Sox2, and c-Myc. After transfection, embryonic fibroblasts were reseeded onto inactivated SNL 76/7 and cultured in a medium supplemented with basic fibroblast growth factor. Flat, compact, primary colonies resembling human iPS colonies were observed. Additionally, primary colonies were more frequently observed in the KnockOut Serum Replacement medium than in the fetal bovine serum (FBS) medium. However, enhanced maintenance and proliferation of iPS-like cells occurred in the FBS medium. These iPS-like cells expressed embryonic stem cell markers, had normal karyotypes, proliferated beyond 45 passages, and differentiated into all three germ layers in vitro. Notably, expression of exogenous Oct3/4, Klf4, and Sox2 was silenced in these cells. However, the iPS-like cells failed to form teratomas. In conclusion, this is the first study to establish and characterize cat iPS-like cells, which can differentiate into different cell types depending on the basic fibroblast growth factor.
Collapse
Affiliation(s)
- Ryoji Kanegi
- Department of Advanced Pathobiology, Graduate School of Veterinary Sciences, Osaka Metropolitan University, Osaka 598-8531, Japan
- Department of Advanced Pathobiology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Osaka 598-8531, Japan
| | - Shingo Hatoya
- Department of Advanced Pathobiology, Graduate School of Veterinary Sciences, Osaka Metropolitan University, Osaka 598-8531, Japan
- Department of Advanced Pathobiology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Osaka 598-8531, Japan
| | - Kazuto Kimura
- Department of Advanced Pathobiology, Graduate School of Veterinary Sciences, Osaka Metropolitan University, Osaka 598-8531, Japan
- Department of Advanced Pathobiology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Osaka 598-8531, Japan
| | - Kyohei Yodoe
- Department of Advanced Pathobiology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Osaka 598-8531, Japan
| | - Toshiya Nishimura
- Department of Advanced Pathobiology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Osaka 598-8531, Japan
| | - Kikuya Sugiura
- Department of Advanced Pathobiology, Graduate School of Veterinary Sciences, Osaka Metropolitan University, Osaka 598-8531, Japan
- Department of Advanced Pathobiology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Osaka 598-8531, Japan
| | - Noritoshi Kawate
- Department of Advanced Pathobiology, Graduate School of Veterinary Sciences, Osaka Metropolitan University, Osaka 598-8531, Japan
- Department of Advanced Pathobiology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Osaka 598-8531, Japan
| | - Toshio Inaba
- Department of Advanced Pathobiology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Osaka 598-8531, Japan
| |
Collapse
|
36
|
Aizawa E, Ozonov EA, Kawamura YK, Dumeau C, Nagaoka S, Kitajima TS, Saitou M, Peters AHFM, Wutz A. Epigenetic regulation limits competence of pluripotent stem cell-derived oocytes. EMBO J 2023; 42:e113955. [PMID: 37850882 PMCID: PMC10690455 DOI: 10.15252/embj.2023113955] [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: 03/07/2023] [Revised: 09/18/2023] [Accepted: 09/18/2023] [Indexed: 10/19/2023] Open
Abstract
Recent studies have reported the differentiation of pluripotent cells into oocytes in vitro. However, the developmental competence of in vitro-generated oocytes remains low. Here, we perform a comprehensive comparison of mouse germ cell development in vitro over all culture steps versus in vivo with the goal to understand mechanisms underlying poor oocyte quality. We show that the in vitro differentiation of primordial germ cells to growing oocytes and subsequent follicle growth is critical for competence for preimplantation development. Systematic transcriptome analysis of single oocytes that were subjected to different culture steps identifies genes that are normally upregulated during oocyte growth to be susceptible for misregulation during in vitro oogenesis. Many misregulated genes are Polycomb targets. Deregulation of Polycomb repression is therefore a key cause and the earliest defect known in in vitro oocyte differentiation. Conversely, structurally normal in vitro-derived oocytes fail at zygotic genome activation and show abnormal acquisition of 5-hydroxymethylcytosine on maternal chromosomes. Our data identify epigenetic regulation at an early stage of oogenesis limiting developmental competence and suggest opportunities for future improvements.
Collapse
Affiliation(s)
- Eishi Aizawa
- Institute of Molecular Health Sciences, Swiss Federal Institute of TechnologyETH ZurichZurichSwitzerland
- RIKEN Center for Biosystems Dynamics ResearchKobeJapan
| | - Evgeniy A Ozonov
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
| | - Yumiko K Kawamura
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
| | - Charles‐Etienne Dumeau
- Institute of Molecular Health Sciences, Swiss Federal Institute of TechnologyETH ZurichZurichSwitzerland
| | - So Nagaoka
- Department of EmbryologyNara Medical UniversityNaraJapan
| | | | - Mitinori Saitou
- Institute for the Advanced Study of Human Biology (ASHBi)Kyoto UniversityKyotoJapan
- Department of Anatomy and Cell Biology, Graduate School of MedicineKyoto UniversityKyotoJapan
- Center for iPS Cell Research and Application (CiRA)Kyoto UniversityKyotoJapan
| | - Antoine HFM Peters
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
- Faculty of SciencesUniversity of BaselBaselSwitzerland
| | - Anton Wutz
- Institute of Molecular Health Sciences, Swiss Federal Institute of TechnologyETH ZurichZurichSwitzerland
| |
Collapse
|
37
|
Chen B, Pei D. Genetic clues to reprogramming power and formation of mouse oocyte. Curr Opin Genet Dev 2023; 83:102110. [PMID: 37722148 DOI: 10.1016/j.gde.2023.102110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/05/2023] [Accepted: 07/29/2023] [Indexed: 09/20/2023]
Abstract
Oocyte features the unique capacity to reprogram not only sperm but also somatic nuclei to totipotency, yet the scarcity of oocytes has hindered the exploration and application of their reprogramming ability. In the meanwhile, the formation of oocytes, which involves extensive intracellular alterations and interactions, has also attracted tremendous interest. This review discusses developmental principles and regulatory mechanisms associated with ooplasm reprogramming and oocyte formation from a genetic perspective, with knowledge derived from mouse models. We also discuss future directions, especially to address the lack of insight into the regulatory networks that shape the identity of female germ cells or drive transitions in their developmental programs.
Collapse
|
38
|
Fair T, Lonergan P. The oocyte: the key player in the success of assisted reproduction technologies. Reprod Fertil Dev 2023; 36:133-148. [PMID: 38064189 DOI: 10.1071/rd23164] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023] Open
Abstract
The ovulation of a mature oocyte at metaphase II of meiosis, with optimal potential to undergo fertilisation by a sperm cell, complete meiosis and sustain the switch to mitotic division, and support early embryo development, involves a protracted and disrupted/delayed series of processes. Many of these are targeted for exploitation in vivo , or recapitulation in vitro , by the livestock industry. Reproductive technologies, including AI, multiple ovulation embryo transfer, ovum pick-up, in vitro embryo production, and oestrus and ovulation synchronisation, offer practitioners and producers the opportunity to produce offspring from genetically valuable dams in much greater numbers than they would normally have in their lifetime, while in vitro oocyte and follicle culture are important platforms for researchers to interrogate the physiological mechanisms driving fertility. The majority of these technologies target the ovarian follicle and the oocyte within; thus, the quality and capability of the recovered oocyte determine the success of the reproductive intervention. Molecular and microscopical technologies have grown exponentially, providing powerful platforms to interrogate the molecular mechanisms which are integral to or affected by ART. The development of the bovine oocyte from its differentiation in the ovary to ovulation is described in the light of its relevance to key aspects of individual interventions, while highlighting the historical timeline.
Collapse
Affiliation(s)
- Trudee Fair
- School of Agriculture and Food Science, University College Dublin, Belfield, Dublin, Ireland
| | - Pat Lonergan
- School of Agriculture and Food Science, University College Dublin, Belfield, Dublin, Ireland
| |
Collapse
|
39
|
Cabezas MR. [Some present and future ethical dilemmas surrounding advancements in in vitro fertilization.]. Salud Colect 2023; 19:e4462. [PMID: 38000000 PMCID: PMC11930338 DOI: 10.18294/sc.2023.4462] [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: 04/14/2023] [Revised: 10/12/2023] [Accepted: 10/26/2023] [Indexed: 11/26/2023] Open
Abstract
The growing field of assisted human reproduction has achieved unimaginable milestones. Its continuous development and the innovations it generates at times pose both ethical and legal dilemmas. This essay aims to elucidate the progressive changes occurring in the realm of the origin of life due to the development of new options and strategies in assisted human reproduction. First, it constructs an interdisciplinary reflection on human nature and the changes society faces from the perspectives of science, ethics, and law, particularly from the perspective of Spain. Second, it provides a brief overview of current or future biomedical techniques in the field of human reproduction. It concludes with a discussion of the need to reflect on the rapid advancement of science in assisted human reproduction.
Collapse
Affiliation(s)
- Marta Reguera Cabezas
- Bióloga, Magíster en Genética y Reproducción Humana Asistida, Unidad de Reproducción Asistida, miembro del Comité de Ética Asistencial, Hospital Universitario Marqués de Valdecilla, Cantabria, España. Hospital Universitario Marqués de ValdecillaUnidad de Reproducción AsistidaComité de Ética AsistencialHospital Universitario Marqués de ValdecillaCantabriaEspaña
| |
Collapse
|
40
|
Sosa E, Mumu SK, Alvarado CC, Wu QY, Roberson I, Espinoza A, Hsu FM, Saito K, Hunt TJ, Faith JE, Lowe MG, DiRusso JA, Clark AT. Reconstituted ovaries self-assemble without an ovarian surface epithelium. Stem Cell Reports 2023; 18:2190-2202. [PMID: 37890483 PMCID: PMC10679655 DOI: 10.1016/j.stemcr.2023.10.001] [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: 07/28/2023] [Revised: 10/01/2023] [Accepted: 10/02/2023] [Indexed: 10/29/2023] Open
Abstract
Three-dimensional (3D) stem cell models of the ovary have the potential to benefit women's reproductive health research. One such model, the reconstituted ovary (rOvary) self-assembles with pluripotent stem cell-derived germ cells creating a 3D ovarian mimic competent to support the differentiation of functional oocytes inside follicles. In this study, we evaluated the cellular composition of the rOvary revealing the capacity to generate multiple follicles surrounded by NR2F2+ stroma cells. However, the rOvary does not develop a surface epithelium, the source of second-wave pre-granulosa cells, or steroidogenic theca. Therefore, the rOvary models represent the self-assembly of activated follicles in a pre-pubertal ovary poised but not yet competent for hormone production.
Collapse
Affiliation(s)
- Enrique Sosa
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Center for Reproductive Science, Health and Education, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Sinthia Kabir Mumu
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Center for Reproductive Science, Health and Education, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Christian C Alvarado
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Center for Reproductive Science, Health and Education, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Qiu Ya Wu
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Center for Reproductive Science, Health and Education, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Isaias Roberson
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Center for Reproductive Science, Health and Education, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Alejandro Espinoza
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Institute for Quantitative and Computational Biosciences - The Collaboratory, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Fei-Man Hsu
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kaori Saito
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Timothy J Hunt
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jared E Faith
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Matthew G Lowe
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jonathan A DiRusso
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Center for Reproductive Science, Health and Education, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Amander T Clark
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Center for Reproductive Science, Health and Education, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
41
|
Appeltant R, Hermes R, Holtze S, Modina SC, Galli C, Bjarkadottir BD, Adeniran BV, Wei X, Swegen A, Hildebrandt TB, Williams SA. The neonatal southern white rhinoceros ovary contains oogonia in germ cell nests. Commun Biol 2023; 6:1049. [PMID: 37848538 PMCID: PMC10582104 DOI: 10.1038/s42003-023-05256-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 08/18/2023] [Indexed: 10/19/2023] Open
Abstract
The northern white rhinoceros is functionally extinct with only two females left. Establishing methods to culture ovarian tissues, follicles, and oocytes to generate eggs will support conservation efforts using in vitro embryo production. To the best of our knowledge, this is the first description of the structure and molecular signature of any rhinoceros, more specifically, we describe the neonatal and adult southern white rhinoceros (Ceratotherium simum simum) ovary; the closest relation of the northern white rhinoceros. Interestingly, all ovaries contain follicles despite advanced age. Analysis of the neonate reveals a population of cells molecularly characterised as mitotically active, pluripotent with germ cell properties. These results indicate that unusually, the neonatal ovary still contains oogonia in germ cell nests at birth, providing an opportunity for fertility preservation. Therefore, utilising ovaries from stillborn and adult rhinoceros can provide cells for advanced assisted reproductive technologies and investigating the neonatal ovaries of other endangered species is crucial for conservation.
Collapse
Affiliation(s)
- Ruth Appeltant
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Women's Centre, Level 3, John Radcliffe Hospital, Oxford, UK
- Gamete Research Centre, Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Robert Hermes
- Leibniz Institute for Zoo and Wildlife Research, Alfred-Kowalke-Str 17, D-10315, Berlin, Germany
| | - Susanne Holtze
- Leibniz Institute for Zoo and Wildlife Research, Alfred-Kowalke-Str 17, D-10315, Berlin, Germany
| | - Silvia Clotilde Modina
- Dipartimento di Medicina Veterinaria, Università degli Studi di Milano, Via dell'Università 6, 26900, Lodi, Italy
| | - Cesare Galli
- Avantea srl, Laboratory of Reproductive Technologies, Via Porcellasco 7/F, 26100, Cremona, Italy
- Fondazione Avantea, 26100, Cremona, Italy
| | - Briet D Bjarkadottir
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Women's Centre, Level 3, John Radcliffe Hospital, Oxford, UK
| | - Babatomisin V Adeniran
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Women's Centre, Level 3, John Radcliffe Hospital, Oxford, UK
| | - Xi Wei
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Women's Centre, Level 3, John Radcliffe Hospital, Oxford, UK
| | - Aleona Swegen
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Women's Centre, Level 3, John Radcliffe Hospital, Oxford, UK
- Priority Research Centre for Reproductive Science, University of Newcastle, Callaghan, 2308, NSW, Australia
| | - Thomas Bernd Hildebrandt
- Dipartimento di Medicina Veterinaria, Università degli Studi di Milano, Via dell'Università 6, 26900, Lodi, Italy
- Freie Universität Berlin, D-14195, Berlin, Germany
| | - Suzannah A Williams
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Women's Centre, Level 3, John Radcliffe Hospital, Oxford, UK.
| |
Collapse
|
42
|
Romualdez-Tan MV. Modelling in vitro gametogenesis using induced pluripotent stem cells: a review. CELL REGENERATION (LONDON, ENGLAND) 2023; 12:33. [PMID: 37843621 PMCID: PMC10579208 DOI: 10.1186/s13619-023-00176-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 08/28/2023] [Indexed: 10/17/2023]
Abstract
In vitro gametogenesis (IVG) has been a topic of great interest in recent years not only because it allows for further exploration of mechanisms of germ cell development, but also because of its prospect for innovative medical applications especially for the treatment of infertility. Elucidation of the mechanisms underlying gamete development in vivo has inspired scientists to attempt to recapitulate the entire process of gametogenesis in vitro. While earlier studies have established IVG methods largely using pluripotent stem cells of embryonic origin, the scarcity of sources for these cells and the ethical issues involved in their use are serious limitations to the progress of IVG research especially in humans. However, with the emergence of induced pluripotent stem cells (iPSCs) due to the revolutionary discovery of dedifferentiation and reprogramming factors, IVG research has progressed remarkably in the last decade. This paper extensively reviews developments in IVG using iPSCs. First, the paper presents key concepts from groundwork studies on IVG including earlier researches demonstrating that IVG methods using embryonic stem cells (ESCs) also apply when using iPSCs. Techniques for the derivation of iPSCs are briefly discussed, highlighting the importance of generating transgene-free iPSCs with a high capacity for germline transmission to improve efficacy when used for IVG. The main part of the paper discusses recent advances in IVG research using iPSCs in various stages of gametogenesis. In addition, current clinical applications of IVG are presented, and potential future applications are discussed. Although IVG is still faced with many challenges in terms of technical issues, as well as efficacy and safety, novel IVG methodologies are emerging, and IVG using iPSCs may usher in the next era of reproductive medicine sooner than expected. This raises both ethical and social concerns and calls for the scientific community to cautiously develop IVG technology to ensure it is not only efficacious but also safe and adheres to social and ethical norms.
Collapse
Affiliation(s)
- Maria Victoria Romualdez-Tan
- Present Address: Repro Optima Center for Reproductive Health, Inc., Ground Floor JRDC Bldg. Osmena Blvd. Capitol Site, Cebu City, 6000, Philippines.
- Cebu Doctors University Hospital, Cebu City, Philippines.
| |
Collapse
|
43
|
Salvatore G, Dolci S, Camaioni A, Klinger FG, De Felici M. Reprogramming Human Female Adipose Mesenchymal Stem Cells into Primordial Germ Cell-Like Cells. Stem Cell Rev Rep 2023; 19:2274-2283. [PMID: 37338786 PMCID: PMC10579115 DOI: 10.1007/s12015-023-10561-x] [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] [Accepted: 05/15/2023] [Indexed: 06/21/2023]
Abstract
In the last two decades, considerable progress has been made in the derivation of mammalian germ cells from pluripotent stem cells such as Embryonic Stem Cells (ESCs) and induced Pluripotent Stem Cells (iPSCs). The pluripotent stem cells are generally first induced into pre-gastrulating endoderm/mesoderm-like status and then specified into putative primordial germ cells (PGCs) termed PGC-like cells (PGCLCs) which possess the potential to generate oocytes and sperms. Adipose-derived mesenchymal stromal cells (ASCs) are multipotent cells, having the capacity to differentiate into cell types such as adipocytes, osteocytes and chondrocytes. Since no information is available about the capability of female human ASCs (hASCs) to generate PGCLCs, we compared protocols to produce such cells from hASCs themselves or from hASC-derived iPSCs. The results showed that, providing pre-induction into a peri-gastrulating endoderm/mesoderm-like status, hASCs can generate PGCLCs. This process, however, shows a lower efficiency than when hASC-derived iPSCs are used as starting cells. Although hASCs possess multipotency and express mesodermal genes, direct induction into PGCLCs resulted less efficient.
Collapse
Affiliation(s)
- Giulia Salvatore
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, Rome, 00133, Italy
| | - Susanna Dolci
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, Rome, 00133, Italy
| | - Antonella Camaioni
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, Rome, 00133, Italy
| | - Francesca Gioia Klinger
- Saint Camillus International University Of Health Sciences, Via di Sant'Alessandro 8, Rome, 00131, Italy.
| | - Massimo De Felici
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, Rome, 00133, Italy.
| |
Collapse
|
44
|
Telfer EE, Grosbois J, Odey YL, Rosario R, Anderson RA. Making a good egg: human oocyte health, aging, and in vitro development. Physiol Rev 2023; 103:2623-2677. [PMID: 37171807 PMCID: PMC10625843 DOI: 10.1152/physrev.00032.2022] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 05/03/2023] [Accepted: 05/06/2023] [Indexed: 05/13/2023] Open
Abstract
Mammalian eggs (oocytes) are formed during fetal life and establish associations with somatic cells to form primordial follicles that create a store of germ cells (the primordial pool). The size of this pool is influenced by key events during the formation of germ cells and by factors that influence the subsequent activation of follicle growth. These regulatory pathways must ensure that the reserve of oocytes within primordial follicles in humans lasts for up to 50 years, yet only approximately 0.1% will ever be ovulated with the rest undergoing degeneration. This review outlines the mechanisms and regulatory pathways that govern the processes of oocyte and follicle formation and later growth, within the ovarian stroma, through to ovulation with particular reference to human oocytes/follicles. In addition, the effects of aging on female reproductive capacity through changes in oocyte number and quality are emphasized, with both the cellular mechanisms and clinical implications discussed. Finally, the details of current developments in culture systems that support all stages of follicle growth to generate mature oocytes in vitro and emerging prospects for making new oocytes from stem cells are outlined.
Collapse
Affiliation(s)
- Evelyn E Telfer
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Johanne Grosbois
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Yvonne L Odey
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Roseanne Rosario
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Richard A Anderson
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
45
|
Mizuta K, Saitou M. Key mechanisms and in vitro reconstitution of fetal oocyte development in mammals. Curr Opin Genet Dev 2023; 82:102091. [PMID: 37556984 DOI: 10.1016/j.gde.2023.102091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/23/2023] [Accepted: 07/01/2023] [Indexed: 08/11/2023]
Abstract
During fetal oocyte development in mammals, germ cells progress through meiotic prophase I to form primordial follicles with pregranulosa cells. The primordial follicles remain dormant until oogenesis resumes during puberty. Studies in mice have elucidated mechanisms governing oogenesis, leading to the successful induction of functional oocytes from mouse pluripotent stem cells in vitro. Based on the in vivo/in vitro knowledge in mice and the histological and transcriptomic evidence for fetal oocyte development in humans and primates, human/primate oocyte-like cells corresponding to the early stage of oocytes in vivo have been successfully induced in vitro. Here, we discuss recent advances in our understanding of the mechanisms of fetal oocyte development in mammals, as well as in in vitro oogenesis.
Collapse
Affiliation(s)
- Ken Mizuta
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan; Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan.
| | - Mitinori Saitou
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan; Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan; Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan.
| |
Collapse
|
46
|
Horer S, Feichtinger M, Rosner M, Hengstschläger M. Pluripotent Stem Cell-Derived In Vitro Gametogenesis and Synthetic Embryos-It Is Never Too Early for an Ethical Debate. Stem Cells Transl Med 2023; 12:569-575. [PMID: 37471266 PMCID: PMC10502567 DOI: 10.1093/stcltm/szad042] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/23/2023] [Indexed: 07/22/2023] Open
Abstract
Recently, 2 branches of the wide area of synthetic biology-in vitro gametogenesis and synthetic embryo development-have gained considerable attention. Rodent induced pluripotent stem cells derived via reprogramming of somatic cells can in vitro be differentiated into gametes to produce fertile offspring. And even synthetic embryos with organ progenitors were generated ex utero entirely from murine pluripotent stem cells. The use of these approaches in basic research, which is rightfully accompanied by an ethical discussion, will allow hitherto unattainable insights into the processes of the beginning of life. There is a broad international consensus that currently the application of these technologies in human-assisted reproduction must be considered to be unsafe and unethical. However, newspaper headlines also addressed the putatively resulting paradigm shift in human reproduction and thereby raised expectations in patients. Due to unsolved biological and technological obstacles, most scientists do not anticipate translation of any of these approaches into human reproductive medicine, if ever, for the next 10 years. Still, whereas the usage of synthetic embryos for reproductive purposes should be banned, in the context of in vitro-derived human gametes it is not too early to initiate the evaluation of the ethical implications, which could still remain assuming all technological hurdles can ever be cleared.
Collapse
Affiliation(s)
- Stefanie Horer
- Institute of Medical Genetics, Center of Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | | | - Margit Rosner
- Institute of Medical Genetics, Center of Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Markus Hengstschläger
- Institute of Medical Genetics, Center of Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
47
|
Cooke CB, Barrington C, Baillie-Benson P, Nichols J, Moris N. Gastruloid-derived primordial germ cell-like cells develop dynamically within integrated tissues. Development 2023; 150:dev201790. [PMID: 37526602 PMCID: PMC10508693 DOI: 10.1242/dev.201790] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/24/2023] [Indexed: 08/02/2023]
Abstract
Primordial germ cells (PGCs) are the early embryonic precursors of gametes - sperm and egg cells. PGC-like cells (PGCLCs) can currently be derived in vitro from pluripotent cells exposed to signalling cocktails and aggregated into large embryonic bodies, but these do not recapitulate the native embryonic environment during PGC formation. Here, we show that mouse gastruloids, a three-dimensional in vitro model of gastrulation, contain a population of gastruloid-derived PGCLCs (Gld-PGCLCs) that resemble early PGCs in vivo. Importantly, the conserved organisation of mouse gastruloids leads to coordinated spatial and temporal localisation of Gld-PGCLCs relative to surrounding somatic cells, even in the absence of specific exogenous PGC-specific signalling or extra-embryonic tissues. In gastruloids, self-organised interactions between cells and tissues, including the endodermal epithelium, enables the specification and subsequent maturation of a pool of Gld-PGCLCs. As such, mouse gastruloids represent a new source of PGCLCs in vitro and, owing to their inherent co-development, serve as a novel model to study the dynamics of PGC development within integrated tissue environments.
Collapse
Affiliation(s)
- Christopher B. Cooke
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
- Abcam, Discovery Drive, Cambridge Biomedical Campus, Cambridge CB2 0AX, UK
| | | | - Peter Baillie-Benson
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Wellcome Trust – MRC Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Tennis Court Road, Cambridge CB2 3EG, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Jennifer Nichols
- Wellcome Trust – MRC Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Tennis Court Road, Cambridge CB2 3EG, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Naomi Moris
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| |
Collapse
|
48
|
Wu J, Shi Y, Yang S, Tang Z, Li Z, Li Z, Zuo J, Ji W, Niu Y. Current state of stem cell research in non-human primates: an overview. MEDICAL REVIEW (2021) 2023; 3:277-304. [PMID: 38235400 PMCID: PMC10790211 DOI: 10.1515/mr-2023-0035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 10/04/2023] [Indexed: 01/19/2024]
Abstract
The remarkable similarity between non-human primates (NHPs) and humans establishes them as essential models for understanding human biology and diseases, as well as for developing novel therapeutic strategies, thereby providing more comprehensive reference data for clinical treatment. Pluripotent stem cells such as embryonic stem cells and induced pluripotent stem cells provide unprecedented opportunities for cell therapies against intractable diseases and injuries. As continue to harness the potential of these biotechnological therapies, NHPs are increasingly being employed in preclinical trials, serving as a pivotal tool to evaluate the safety and efficacy of these interventions. Here, we review the recent advancements in the fundamental research of stem cells and the progress made in studies involving NHPs.
Collapse
Affiliation(s)
- Junmo Wu
- Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan Province, China
| | - Yuxi Shi
- Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan Province, China
| | - Shanshan Yang
- Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan Province, China
| | - Zengli Tang
- Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan Province, China
| | - Zifan Li
- Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan Province, China
| | - Zhuoyao Li
- Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan Province, China
| | - Jiawei Zuo
- Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan Province, China
| | - Weizhi Ji
- Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan Province, China
| | - Yuyu Niu
- Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan Province, China
| |
Collapse
|
49
|
Rosner M, Horer S, Feichtinger M, Hengstschläger M. Multipotent fetal stem cells in reproductive biology research. Stem Cell Res Ther 2023; 14:157. [PMID: 37287077 DOI: 10.1186/s13287-023-03379-4] [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: 01/06/2023] [Accepted: 05/16/2023] [Indexed: 06/09/2023] Open
Abstract
Due to the limited accessibility of the in vivo situation, the scarcity of the human tissue, legal constraints, and ethical considerations, the underlying molecular mechanisms of disorders, such as preeclampsia, the pathological consequences of fetomaternal microchimerism, or infertility, are still not fully understood. And although substantial progress has already been made, the therapeutic strategies for reproductive system diseases are still facing limitations. In the recent years, it became more and more evident that stem cells are powerful tools for basic research in human reproduction and stem cell-based approaches moved into the center of endeavors to establish new clinical concepts. Multipotent fetal stem cells derived from the amniotic fluid, amniotic membrane, chorion leave, Wharton´s jelly, or placenta came to the fore because they are easy to acquire, are not associated with ethical concerns or covered by strict legal restrictions, and can be banked for autologous utilization later in life. Compared to adult stem cells, they exhibit a significantly higher differentiation potential and are much easier to propagate in vitro. Compared to pluripotent stem cells, they harbor less mutations, are not tumorigenic, and exhibit low immunogenicity. Studies on multipotent fetal stem cells can be invaluable to gain knowledge on the development of dysfunctional fetal cell types, to characterize the fetal stem cells migrating into the body of a pregnant woman in the context of fetomaternal microchimerism, and to obtain a more comprehensive picture of germ cell development in the course of in vitro differentiation experiments. The in vivo transplantation of fetal stem cells or their paracrine factors can mediate therapeutic effects in preeclampsia and can restore reproductive organ functions. Together with the use of fetal stem cell-derived gametes, such strategies could once help individuals, who do not develop functional gametes, to conceive genetically related children. Although there is still a long way to go, these developments regarding the usage of multipotent fetal stem cells in the clinic should continuously be accompanied by a wide and detailed ethical discussion.
Collapse
Affiliation(s)
- Margit Rosner
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria
| | - Stefanie Horer
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria
| | | | - Markus Hengstschläger
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Strasse 10, 1090, Vienna, Austria.
| |
Collapse
|
50
|
Wu GMJ, Chen ACH, Yeung WSB, Lee YL. Current progress on in vitro differentiation of ovarian follicles from pluripotent stem cells. Front Cell Dev Biol 2023; 11:1166351. [PMID: 37325555 PMCID: PMC10267358 DOI: 10.3389/fcell.2023.1166351] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/22/2023] [Indexed: 06/17/2023] Open
Abstract
Mammalian female reproduction requires a functional ovary. Competence of the ovary is determined by the quality of its basic unit-ovarian follicles. A normal follicle consists of an oocyte enclosed within ovarian follicular cells. In humans and mice, the ovarian follicles are formed at the foetal and the early neonatal stage respectively, and their renewal at the adult stage is controversial. Extensive research emerges recently to produce ovarian follicles in-vitro from different species. Previous reports demonstrated the differentiation of mouse and human pluripotent stem cells into germline cells, termed primordial germ cell-like cells (PGCLCs). The germ cell-specific gene expressions and epigenetic features including global DNA demethylation and histone modifications of the pluripotent stem cells-derived PGCLCs were extensively characterized. The PGCLCs hold potential for forming ovarian follicles or organoids upon cocultured with ovarian somatic cells. Intriguingly, the oocytes isolated from the organoids could be fertilized in-vitro. Based on the knowledge of in-vivo derived pre-granulosa cells, the generation of these cells from pluripotent stem cells termed foetal ovarian somatic cell-like cells was also reported recently. Despite successful in-vitro folliculogenesis from pluripotent stem cells, the efficiency remains low, mainly due to the lack of information on the interaction between PGCLCs and pre-granulosa cells. The establishment of in-vitro pluripotent stem cell-based models paves the way for understanding the critical signalling pathways and molecules during folliculogenesis. This article aims to review the developmental events during in-vivo follicular development and discuss the current progress of generation of PGCLCs, pre-granulosa and theca cells in-vitro.
Collapse
Affiliation(s)
- Genie Min Ju Wu
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, The University of Hong Kong, Hong Kong, China
| | - Andy Chun Hang Chen
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, The University of Hong Kong, Hong Kong, China
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong—Shenzhen Hospital, Shenzhen, China
- Centre for Translational Stem Cell Biology, The Hong Kong Science and Technology Park, Hong Kong, China
| | - William Shu Biu Yeung
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, The University of Hong Kong, Hong Kong, China
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong—Shenzhen Hospital, Shenzhen, China
- Centre for Translational Stem Cell Biology, The Hong Kong Science and Technology Park, Hong Kong, China
| | - Yin Lau Lee
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, The University of Hong Kong, Hong Kong, China
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong—Shenzhen Hospital, Shenzhen, China
- Centre for Translational Stem Cell Biology, The Hong Kong Science and Technology Park, Hong Kong, China
| |
Collapse
|