1
|
Wang G, Korody ML, Brändl B, Hernandez-Toro CJ, Rohrandt C, Hong K, Pang AWC, Lee J, Migliorelli G, Stanke M, Ford SM, Pollmann I, Houck ML, Lewin HA, Lear TL, Ryder OA, Meissner A, Loring JF, Müller FJ. Genomic map of the functionally extinct northern white rhinoceros ( Ceratotherium simum cottoni). Proc Natl Acad Sci U S A 2025; 122:e2401207122. [PMID: 40359041 DOI: 10.1073/pnas.2401207122] [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: 03/04/2024] [Accepted: 03/20/2025] [Indexed: 05/15/2025] Open
Abstract
The northern white rhinoceros (NWR; Ceratotherium simum cottoni) is functionally extinct, with only two nonreproductive females alive. Efforts to rescue the NWR from its inevitable demise have inspired the exploration of unconventional conservation methods, including the development of induced pluripotent stem cells (iPSCs) for the in vitro generation of artificial gametes. The integrity of iPSC genomes is critical for in vitro gametogenesis to be used for assisted reproductive technologies using NWR iPSCs. We generated a chromosome-level NWR reference genome that meets or exceeds the metrics proposed by the Vertebrate Genome Project, using complementary sequencing and mapping methods. The genome represents 40 autosomes, an X and a partially resolved Y chromosome, and the mitochondrial genome. Using comparative FISH mapping, we confirmed a general gene order conservation between the NWR and horse genomes. We aligned the NWR genome with that of the southern white rhinoceros (SWR; Ceratotherium simum simum), a population that has been physically separated from the NWR for tens of thousands of years, and we found that the two subspecies are very similar on the chromosome level. Comparing long-read data from NWR iPSC lines and the fibroblast cultures used for reprogramming, we identified copy number variations that were likely to have been introduced during in vitro iPSC expansion. The NWR reference genome allows for efficient, rapid, and accurate assessment of the genomic integrity of iPSC lines to direct their differentiation. This will assist in strategies to rescue the NWR through extraordinary measures like cloning and the generation of embryos from iPSC-derived gametes.
Collapse
Affiliation(s)
- Gaojianyong Wang
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin 14195, Germany
- Department of Psychiatry and Psychotherapy, Christian-Albrechts Universität, Kiel 24105, Germany
- Zentrum für Integrative Psychiatrie, University Hospital Schleswig-Holstein, Kiel 24105, Germany
| | | | - Björn Brändl
- Department of Psychiatry and Psychotherapy, Christian-Albrechts Universität, Kiel 24105, Germany
- Zentrum für Integrative Psychiatrie, University Hospital Schleswig-Holstein, Kiel 24105, Germany
| | | | - Christian Rohrandt
- Department of Psychiatry and Psychotherapy, Christian-Albrechts Universität, Kiel 24105, Germany
- Zentrum für Integrative Psychiatrie, University Hospital Schleswig-Holstein, Kiel 24105, Germany
- Institute for Communications Technologies and Embedded Systems, Kiel University of Applied Sciences, Kiel 24149, Germany
| | - Karl Hong
- Bionano Genomics Inc, San Diego CA, 92121
| | | | - Joyce Lee
- Bionano Genomics Inc, San Diego CA, 92121
| | - Giovanna Migliorelli
- Institute of Mathematics and Computer Science, and Center for Functional Genomics of Microbes, University of Greifswald, Greifswald 17489, Germany
| | - Mario Stanke
- Institute of Mathematics and Computer Science, and Center for Functional Genomics of Microbes, University of Greifswald, Greifswald 17489, Germany
| | - Sarah M Ford
- San Diego Zoo Wildlife Alliance, Escondido, CA, 92027
- Department of Ecology and Evolutionary Biology, University of California, Santa Cruz, CA 95060
| | - Iris Pollmann
- Department of Psychiatry and Psychotherapy, Christian-Albrechts Universität, Kiel 24105, Germany
- Zentrum für Integrative Psychiatrie, University Hospital Schleswig-Holstein, Kiel 24105, Germany
| | | | - Harris A Lewin
- The Genome Center, University of California, Davis, CA 95616
- Department of Evolution and Ecology, University of California, Davis, CA 95616
- John Muir Institute for the Environment, University of California, Davis, CA 95616
| | - Teri L Lear
- Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, KY 40546
| | | | - Alexander Meissner
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin 14195, Germany
| | | | - Franz-Josef Müller
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin 14195, Germany
- Department of Psychiatry and Psychotherapy, Christian-Albrechts Universität, Kiel 24105, Germany
- Zentrum für Integrative Psychiatrie, University Hospital Schleswig-Holstein, Kiel 24105, Germany
| |
Collapse
|
2
|
Zhao Z, Wu Y, Cheng F, Wang Z, Geng Q, Niu Y, Zuo Q, Zhang Y. High levels of histone acetylation modifications promote the formation of PGCs. Poult Sci 2025; 104:104763. [PMID: 39798283 PMCID: PMC11954803 DOI: 10.1016/j.psj.2024.104763] [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: 08/01/2024] [Revised: 12/28/2024] [Accepted: 12/31/2024] [Indexed: 01/15/2025] Open
Abstract
This study investigates the role of histone acetylation in the differentiation of chicken embryonic stem cells (ESCs) into primordial germ cells (PGCs). Transcriptomic sequencing was used to analyze differentially expressed genes during this differentiation process, with functional annotation identifying genes associated with histone acetylation. To explore the role of acetylation, acetate and an acetyltransferase inhibitor (ANAC) were added to the ESCs induction medium. Transcriptomic analysis revealed that during ESCs differentiation into PGCs, genes involved in histone acetyltransferase activity were upregulated, while those associated with histone deacetylase activity were downregulated. Functional enrichment analysis indicated these genes are involved in pathways critical for germ cell differentiation, underscoring their importance in avian reproductive biology. Quantitative real-time PCR (qRT-PCR) confirmed significant differential expression of HAT8 and HDAC10 between ESCs and PGCs (P < 0.01). The acetate treatment group exhibited a significantly higher number of embryoid bodies and elevated expression levels of CVH, C-KIT, and NANOS3 compared to the ANAC group (P < 0.01). Furthermore, indirect immunofluorescence and flow cytometry demonstrated a significantly higher proportion of DDX4-positive cells in the acetate group (P < 0.01). These findings provide preliminary evidence that histone acetylation regulates chicken PGCs formation, offering a theoretical framework for the epigenetic induction of PGCs in vitro. This study enhances our understanding of the molecular mechanisms underlying PGCs development in poultry and contributes to advancements in avian reproductive technologies and genetic conservation.
Collapse
Affiliation(s)
- Ziduo Zhao
- College of Animal Science and Technology, Yangzhou University, Jiangsu Province Key Laboratory of Animal Breeding and Molecular Design, Yangzhou 225009 Jiangsu, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009 Jiangsu, PR China
| | - Yuhui Wu
- College of Animal Science and Technology, Yangzhou University, Jiangsu Province Key Laboratory of Animal Breeding and Molecular Design, Yangzhou 225009 Jiangsu, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009 Jiangsu, PR China
| | - Fufu Cheng
- College of Animal Science and Technology, Yangzhou University, Jiangsu Province Key Laboratory of Animal Breeding and Molecular Design, Yangzhou 225009 Jiangsu, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009 Jiangsu, PR China
| | - Zhe Wang
- College of Animal Science and Technology, Yangzhou University, Jiangsu Province Key Laboratory of Animal Breeding and Molecular Design, Yangzhou 225009 Jiangsu, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009 Jiangsu, PR China
| | - Qingqing Geng
- College of Animal Science and Technology, Yangzhou University, Jiangsu Province Key Laboratory of Animal Breeding and Molecular Design, Yangzhou 225009 Jiangsu, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009 Jiangsu, PR China
| | - Yingjie Niu
- College of Animal Science and Technology, Yangzhou University, Jiangsu Province Key Laboratory of Animal Breeding and Molecular Design, Yangzhou 225009 Jiangsu, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009 Jiangsu, PR China
| | - Qisheng Zuo
- College of Animal Science and Technology, Yangzhou University, Jiangsu Province Key Laboratory of Animal Breeding and Molecular Design, Yangzhou 225009 Jiangsu, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009 Jiangsu, PR China
| | - Yani Zhang
- College of Animal Science and Technology, Yangzhou University, Jiangsu Province Key Laboratory of Animal Breeding and Molecular Design, Yangzhou 225009 Jiangsu, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009 Jiangsu, PR China.
| |
Collapse
|
3
|
Korody ML, Hildebrandt TB. Progress Toward Genetic Rescue of the Northern White Rhinoceros ( Ceratotherium simum cottoni). Annu Rev Anim Biosci 2025; 13:483-505. [PMID: 39531386 DOI: 10.1146/annurev-animal-111523-102158] [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: 11/16/2024]
Abstract
The northern white rhinoceros (NWR) is functionally extinct, with only two nonreproductive females remaining. However, because of the foresight of scientists, cryopreserved cells and reproductive tissues may aid in the recovery of this species. An ambitious program of natural and artificial gametes and in vitro embryo generation was first outlined in 2015, and many of the proposed steps have been achieved. Multiple induced pluripotent stem cell lines have been established, primordial germ cell-like cells have been generated, oocytes have been collected from the remaining females, blastocysts have been cryopreserved, and the closely related southern white rhinoceros (SWR) is being established as a surrogate. Recently, the first successful embryo transfer in SWR demonstrated that embryos can be generated by in vitro fertilization and cryopreserved. We explore progress to date in using advanced cellular technologies to save the NWR and highlight the necessary next steps to ensure a viable population for reintroduction. We roll out a holistic rescue approach for a charismatic megavertebrate that includes the most advanced cellular technologies, which can provide a blueprint for other critically endangered mammals. We also provide a detailed discussion of the remaining questions in such an upgraded conservation program.
Collapse
Affiliation(s)
- Marisa L Korody
- San Diego Zoo Wildlife Alliance, Escondido, California, USA;
| | - Thomas B Hildebrandt
- Faculty of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
- Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany;
| |
Collapse
|
4
|
Marx V. Can stem cells save the animals? Nat Methods 2025; 22:8-12. [PMID: 39806061 DOI: 10.1038/s41592-024-02577-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
|
5
|
Biasetti P, Mercugliano E, Schrade L, Spiriti MM, Göritz F, Holtze S, Seet S, Galli C, Stejskal J, Colleoni S, Čižmár D, Simone R, Hildebrandt TB, de Mori B. Ethical assessment of genome resource banking (GRB) in wildlife conservation. Cryobiology 2024; 117:104956. [PMID: 39181526 DOI: 10.1016/j.cryobiol.2024.104956] [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: 06/23/2024] [Revised: 07/27/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024]
Abstract
Genome Resources Banks (GRBs) represent vital repositories for the systematic collection, storage, and management of genetic material across various taxa, with a primary objective of safeguarding genetic diversity for research and practical applications. Alongside the development of assisted reproductive techniques (ART), GRBs have evolved into indispensable tools in conservation, offering opportunities for species preservation, mitigating inbreeding risks, and facilitating genetic management across fragmented populations. By preserving genetic information in a suspended state, GRBs serve as backups against population vulnerabilities, potentially aiding in the restoration of endangered species and extending their genetic lifespan. While evidence demonstrates the efficacy of GRBs, ethical considerations surrounding biobanking procedures for wildlife conservation remain largely unexplored. In this article, we will discuss possible ethical issues related to GRBs and the need to ethically monitor biobanking procedures in wildlife conservation. We will then propose a methodological tool, ETHAS, already in use for the ethical self-assessment of assisted reproduction techniques, to assess also biobanking procedures. ETHAS can make it possible to monitor a GRB from its design phase to its actual operation, helping to build biobanking procedures that meet high ethical standards.
Collapse
Affiliation(s)
- Pierfrancesco Biasetti
- Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany; Ethics Laboratory for Veterinary Medicine, Conservation and Animal Welfare, Padua University, Padua, Italy.
| | - Elena Mercugliano
- Ethics Laboratory for Veterinary Medicine, Conservation and Animal Welfare, Padua University, Padua, Italy; Department of Comparative Biomedicine and Food Science, Padua University, Padua, Italy
| | - Lisa Schrade
- Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany
| | - Maria Michela Spiriti
- Ethics Laboratory for Veterinary Medicine, Conservation and Animal Welfare, Padua University, Padua, Italy
| | - Frank Göritz
- Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany
| | - Susanne Holtze
- Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany
| | - Steven Seet
- Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany
| | | | | | | | - Daniel Čižmár
- Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany
| | - Raffaella Simone
- Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany
| | | | - Barbara de Mori
- Ethics Laboratory for Veterinary Medicine, Conservation and Animal Welfare, Padua University, Padua, Italy; Department of Comparative Biomedicine and Food Science, Padua University, Padua, Italy.
| |
Collapse
|
6
|
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
|
7
|
Hildebrandt TB, Holtze S. Advanced assisted reproduction technologies in endangered mammalian species. Reprod Domest Anim 2024; 59 Suppl 3:e14700. [PMID: 39396875 DOI: 10.1111/rda.14700] [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: 05/14/2024] [Revised: 07/12/2024] [Accepted: 07/24/2024] [Indexed: 10/15/2024]
Abstract
A new synergistic approach of classical conservation strategies combined with advanced assisted reproduction technologies (aART) allows for protection and rescue of endangered keystone species at the brink of extinction, which can help to safeguard complex ecosystems. Reproduction biology and management in mammal species is not only challenging in regards to their diverging sizes, anatomy, and often unknown physiology; it also requires customized training or chemical restraint protocols for safe handling. Besides these general challenges, there are several new assisted reproduction techniques (ART) specifically tailored to critically endangered mammals. The current portfolio of ART in these mammalian taxa is ranging from sexual cycle characterization and manipulation, semen collection and cryopreservation, artificial insemination, biobanking of living cells, oocyte collection, in vitro fertilization (IVF), and embryo production, embryo transfer as well as stem cell-derived in vitro gametogenesis for generating gametes in culture. The article covers advanced assisted reproduction technologies (aART), success and challenges, as well as ethical implications.
Collapse
Affiliation(s)
- Thomas Bernd Hildebrandt
- Leibniz Institute for Zoo and Wildlife Research (IZW), Berlin, Germany
- Faculty Veterinary Medicine, Freie Universitaet Berlin, Berlin, Germany
| | - Susanne Holtze
- Leibniz Institute for Zoo and Wildlife Research (IZW), Berlin, Germany
| |
Collapse
|
8
|
Liu Y, Zhang S, Zou G, An J, Li Y, Lin D, Wang D, Li Y, Chen J, Feng T, Li H, Chen Y, Zhang M, Kumar M, Wang L, Hou R, Liu J. Generation and characterization of giant panda induced pluripotent stem cells. SCIENCE ADVANCES 2024; 10:eadn7724. [PMID: 39303041 DOI: 10.1126/sciadv.adn7724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 08/14/2024] [Indexed: 09/22/2024]
Abstract
The giant panda (Ailuropoda melanoleuca) stands as a flagship and umbrella species, symbolizing global biodiversity. While traditional assisted reproductive technology faces constraints in safeguarding the genetic diversity of giant pandas, induced pluripotent stem cells (iPSCs) known for their capacity to differentiate into diverse cells types, including germ cells, present a transformative potential for conservation of endangered animals. In this study, primary fibroblast cells were isolated from the giant panda, and giant panda iPSCs (GPiPSCs) were generated using a non-integrating episomal vector reprogramming method. Characterization of these GPiPSCs revealed their state of primed pluripotency and demonstrated their potential for differentiation. Furthermore, we innovatively formulated a species-specific chemically defined FACL medium and unraveled the intricate signaling pathway networks responsible for maintaining the pluripotency and fostering cell proliferation of GPiPSCs. This study provides key insights into rare species iPSCs, offering materials for panda characteristics research and laying the groundwork for in vitro giant panda gamete generation, potentially aiding endangered species conservation.
Collapse
Affiliation(s)
- Yuliang Liu
- Chengdu Research Base of Giant Panda Breeding, Sichuan Province, Chengdu 610081, China
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Sichuan Province, Chengdu 610081, China
| | - Shihao Zhang
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Gaoyang Zou
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory at GIBH, Guangzhou 510530, China
| | - Junhui An
- Chengdu Research Base of Giant Panda Breeding, Sichuan Province, Chengdu 610081, China
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Sichuan Province, Chengdu 610081, China
| | - Yuan Li
- Chengdu Research Base of Giant Panda Breeding, Sichuan Province, Chengdu 610081, China
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Sichuan Province, Chengdu 610081, China
| | - Danni Lin
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Donghui Wang
- Chengdu Research Base of Giant Panda Breeding, Sichuan Province, Chengdu 610081, China
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Sichuan Province, Chengdu 610081, China
| | - Yan Li
- Chengdu Research Base of Giant Panda Breeding, Sichuan Province, Chengdu 610081, China
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Sichuan Province, Chengdu 610081, China
| | - Jiasong Chen
- Chengdu Research Base of Giant Panda Breeding, Sichuan Province, Chengdu 610081, China
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Sichuan Province, Chengdu 610081, China
| | - Tongying Feng
- Chengdu Research Base of Giant Panda Breeding, Sichuan Province, Chengdu 610081, China
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Sichuan Province, Chengdu 610081, China
| | - Hongyan Li
- Chengdu Research Base of Giant Panda Breeding, Sichuan Province, Chengdu 610081, China
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Sichuan Province, Chengdu 610081, China
| | - Yijiao Chen
- Chengdu Research Base of Giant Panda Breeding, Sichuan Province, Chengdu 610081, China
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Sichuan Province, Chengdu 610081, China
| | - Mingyue Zhang
- Chengdu Research Base of Giant Panda Breeding, Sichuan Province, Chengdu 610081, China
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Sichuan Province, Chengdu 610081, China
| | - Manish Kumar
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Luqin Wang
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Rong Hou
- Chengdu Research Base of Giant Panda Breeding, Sichuan Province, Chengdu 610081, China
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Sichuan Province, Chengdu 610081, China
| | - Jing Liu
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory at GIBH, Guangzhou 510530, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, P.R.China
| |
Collapse
|
9
|
Galli C, Lazzari G. 40 years of AETE: the contribution of scientists and practitioners to the progress of reproductive biotechnologies in Europe. Anim Reprod 2024; 21:e20240061. [PMID: 39286367 PMCID: PMC11404877 DOI: 10.1590/1984-3143-ar2024-0061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 06/20/2024] [Indexed: 09/19/2024] Open
Abstract
This conference celebrates the 40th anniversary of AETE. Over the past 40 years, AETE has served as a forum for scientists, practitioners, and students working in assisted animal reproduction in livestock species. AETE conferences have reflected developments in the field, from basic to applied science, as well as regulatory changes in assisted animal reproduction practices. Europe has led the way in these developments for many years, progressing from artificial insemination, embryo transfer, and cryopreservation to semen sexing, in vitro production of embryos, cloning by nuclear transfer, genomic selection, and the rescue of highly endangered species. These significant contributions were made possible by the support of funding agencies, both at the national and European levels, promoting cooperation between scientists and practitioners. Assisted reproduction, and animal breeding more generally, face opposition from various groups, including animal rights activists, vegetarians, proponents of organic farming, environmentalists, certain political parties, and increasing regulatory burdens. These challenges seriously affect funding for scientific research, the work of practitioners, and the breeding industry as a whole. It is crucial to invest time and resources in communication to remind the public, politicians, and regulators of the achievements in this field and the contributions made to the food supply chain and the care of the rural and natural environment.
Collapse
Affiliation(s)
- Cesare Galli
- Avantea and Fondazione Avantea Onlus, Cremona, Italy
| | | |
Collapse
|
10
|
Mastromonaco G. 40 'wild' years: the current reality and future potential of assisted reproductive technologies in wildlife species. Anim Reprod 2024; 21:e20240049. [PMID: 39286364 PMCID: PMC11404876 DOI: 10.1590/1984-3143-ar2024-0049] [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: 04/18/2024] [Accepted: 06/25/2024] [Indexed: 09/19/2024] Open
Abstract
Over the past 40 years, assisted reproductive technologies (ARTs) have grown significantly in scale and innovation, from the bovine embryo industry's shift from in vivo derived to in vitro produced embryos and the development of somatic cell-based approaches for embryo production. Domestic animal models have been instrumental in the development of ARTs for wildlife species in support of the One Plan Approach to species conservation that integrates in situ and ex situ population management strategies. While ARTs are not the sole solution to the biodiversity crisis, they can offer opportunities to maintain, and even improve, the genetic composition of the captive and wild gene pools over time. This review focuses on the application of sperm and embryo technologies (artificial insemination and multiple ovulation/in vitro produced embryo transfer, respectively) in wildlife species, highlighting impactful cases in which significant progress or innovation has transpired. One of the key messages following decades of efforts in this field is the importance of collaboration between researchers and practitioners from zoological, academic, governmental, and private sectors.
Collapse
|
11
|
Vasan R, Ferrante AJ, Borensztejn A, Frick CL, Gaudreault N, Mogre SS, Morris B, Pires GG, Rafelski SM, Theriot JA, Viana MP. Interpretable representation learning for 3D multi-piece intracellular structures using point clouds. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.25.605164. [PMID: 39091871 PMCID: PMC11291148 DOI: 10.1101/2024.07.25.605164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
A key challenge in understanding subcellular organization is quantifying interpretable measurements of intracellular structures with complex multi-piece morphologies in an objective, robust and generalizable manner. Here we introduce a morphology-appropriate representation learning framework that uses 3D rotation invariant autoencoders and point clouds. This framework is used to learn representations of complex multi-piece morphologies that are independent of orientation, compact, and easy to interpret. We apply our framework to intracellular structures with punctate morphologies (e.g. DNA replication foci) and polymorphic morphologies (e.g. nucleoli). We systematically compare our framework to image-based autoencoders across several intracellular structure datasets, including a synthetic dataset with pre-defined rules of organization. We explore the trade-offs in the performance of different models by performing multi-metric benchmarking across efficiency, generative capability, and representation expressivity metrics. We find that our framework, which embraces the underlying morphology of multi-piece structures, facilitates the unsupervised discovery of sub-clusters for each structure. We show how our approach can also be applied to phenotypic profiling using a dataset of nucleolar images following drug perturbations. We implement and provide all representation learning models using CytoDL, a python package for flexible and configurable deep learning experiments.
Collapse
Affiliation(s)
- Ritvik Vasan
- Allen Institute for Cell Science, Seattle, WA, USA
| | | | | | | | | | | | | | | | | | - Julie A Theriot
- Department of Biology and Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | | |
Collapse
|
12
|
Wang Z, Gong W, Yao Z, Jin K, Niu Y, Li B, Zuo Q. Mechanisms of Embryonic Stem Cell Pluripotency Maintenance and Their Application in Livestock and Poultry Breeding. Animals (Basel) 2024; 14:1742. [PMID: 38929361 PMCID: PMC11201147 DOI: 10.3390/ani14121742] [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: 05/09/2024] [Revised: 05/31/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Embryonic stem cells (ESCs) are remarkably undifferentiated cells that originate from the inner cell mass of the blastocyst. They possess the ability to self-renew and differentiate into multiple cell types, making them invaluable in diverse applications such as disease modeling and the creation of transgenic animals. In recent years, as agricultural practices have evolved from traditional to biological breeding, it has become clear that pluripotent stem cells (PSCs), either ESCs or induced pluripotent stem cells (iPSCs), are optimal for continually screening suitable cellular materials. However, the technologies for long-term in vitro culture or establishment of cell lines for PSCs in livestock are still immature, and research progress is uneven, which poses challenges for the application of PSCs in various fields. The establishment of a robust in vitro system for these cells is critically dependent on understanding their pluripotency maintenance mechanisms. It is believed that the combined effects of pluripotent transcription factors, pivotal signaling pathways, and epigenetic regulation contribute to maintaining their pluripotent state, forming a comprehensive regulatory network. This article will delve into the primary mechanisms underlying the maintenance of pluripotency in PSCs and elaborate on the applications of PSCs in the field of livestock.
Collapse
Affiliation(s)
- Ziyu Wang
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; (Z.W.); (W.G.); (Z.Y.); (K.J.); (Y.N.); (B.L.)
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Wei Gong
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; (Z.W.); (W.G.); (Z.Y.); (K.J.); (Y.N.); (B.L.)
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Zeling Yao
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; (Z.W.); (W.G.); (Z.Y.); (K.J.); (Y.N.); (B.L.)
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Kai Jin
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; (Z.W.); (W.G.); (Z.Y.); (K.J.); (Y.N.); (B.L.)
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Yingjie Niu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; (Z.W.); (W.G.); (Z.Y.); (K.J.); (Y.N.); (B.L.)
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Bichun Li
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; (Z.W.); (W.G.); (Z.Y.); (K.J.); (Y.N.); (B.L.)
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Qisheng Zuo
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; (Z.W.); (W.G.); (Z.Y.); (K.J.); (Y.N.); (B.L.)
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
13
|
Schrade L, Mah N, Bandrowski A, Chen Y, Dewender J, Diecke S, Hiepen C, Lancaster MA, Marques-Bonet T, Martinez S, Mueller SC, Navara C, Prigione A, Seltmann S, Sochacki J, Sutcliffe MA, Zywitza V, Hildebrandt TB, Kurtz A. A Standardized Nomenclature Design for Systematic Referencing and Identification of Animal Cellular Material. Animals (Basel) 2024; 14:1541. [PMID: 38891588 PMCID: PMC11171381 DOI: 10.3390/ani14111541] [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: 03/28/2024] [Revised: 05/15/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
The documentation, preservation and rescue of biological diversity increasingly uses living biological samples. Persistent associations between species, biosamples, such as tissues and cell lines, and the accompanying data are indispensable for using, exchanging and benefiting from these valuable materials. Explicit authentication of such biosamples by assigning unique and robust identifiers is therefore required to allow for unambiguous referencing, avoid identification conflicts and maintain reproducibility in research. A predefined nomenclature based on uniform rules would facilitate this process. However, such a nomenclature is currently lacking for animal biological material. We here present a first, standardized, human-readable nomenclature design, which is sufficient to generate unique and stable identifying names for animal cellular material with a focus on wildlife species. A species-specific human- and machine-readable syntax is included in the proposed standard naming scheme, allowing for the traceability of donated material and cultured cells, as well as data FAIRification. Only when it is consistently applied in the public domain, as publications and inter-institutional samples and data are exchanged, distributed and stored centrally, can the risks of misidentification and loss of traceability be mitigated. This innovative globally applicable identification system provides a standard for a sustainable structure for the long-term storage of animal bio-samples in cryobanks and hence facilitates current as well as future species conservation and biomedical research.
Collapse
Affiliation(s)
- Lisa Schrade
- Fraunhofer Institute for Biomedical Engineering (IBMT), 66280 Sulzbach, Germany
- Department of Reproduction Management, Leibniz Institute for Zoo and Wildlife Research (IZW), 10315 Berlin, Germany
| | - Nancy Mah
- Fraunhofer Institute for Biomedical Engineering (IBMT), 66280 Sulzbach, Germany
| | - Anita Bandrowski
- Department of Neuroscience, FAIR Data Informatics Lab, University of California San Diego, San Diego, CA 92093, USA
- SciCrunch Inc., San Diego, CA 92192, USA
| | - Ying Chen
- Fraunhofer Institute for Biomedical Engineering (IBMT), 66280 Sulzbach, Germany
| | - Johannes Dewender
- Fraunhofer Institute for Biomedical Engineering (IBMT), 66280 Sulzbach, Germany
| | - Sebastian Diecke
- Technology Platform Pluripotent Stem Cells, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Christian Hiepen
- Fraunhofer Institute for Biomedical Engineering (IBMT), 66280 Sulzbach, Germany
| | - Madeline A. Lancaster
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| | - Tomas Marques-Bonet
- Institute of Evolutionary Biology, Pompeu Fabra University—Spanish National Research Council, ICREA, 08003 Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
- Centro Nacional de Analisis Genomico (CNAG), 08028 Barcelona, Spain
- Catalan Institute of Palaeontology Miquel Crusafont, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Sira Martinez
- Institute of Evolutionary Biology, Pompeu Fabra University—Spanish National Research Council, ICREA, 08003 Barcelona, Spain
- European Molecular Biology Laboratory (EMBL) Barcelona, 08003 Barcelona, Spain
| | - Sabine C. Mueller
- Fraunhofer Institute for Biomedical Engineering (IBMT), 66280 Sulzbach, Germany
| | - Christopher Navara
- San Antonio Cellular Therapeutics Institute, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Alessandro Prigione
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Duesseldorf University Hospital, Medical Faculty, Heinrich Heine University, 40225 Duesseldorf, Germany
| | - Stefanie Seltmann
- Fraunhofer Institute for Biomedical Engineering (IBMT), 66280 Sulzbach, Germany
| | - Jaroslaw Sochacki
- European Molecular Biology Laboratory (EMBL) Barcelona, 08003 Barcelona, Spain
| | | | - Vera Zywitza
- Technology Platform Pluripotent Stem Cells, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Thomas B. Hildebrandt
- Department of Reproduction Management, Leibniz Institute for Zoo and Wildlife Research (IZW), 10315 Berlin, Germany
- Faculty of Veterinary Medicine, Free University of Berlin, 14163 Berlin, Germany
| | - Andreas Kurtz
- Fraunhofer Institute for Biomedical Engineering (IBMT), 66280 Sulzbach, Germany
- Berlin Institute of Health (BIH), Center for Regenerative Therapies (BCRT), 13353 Berlin, Germany
| |
Collapse
|
14
|
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
|
15
|
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
|
16
|
Sakaguchi K. In Vitro Growth of Mammalian Follicles and Oocytes. Animals (Basel) 2024; 14:1355. [PMID: 38731360 PMCID: PMC11083657 DOI: 10.3390/ani14091355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 04/10/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024] Open
Abstract
Mammalian ovaries contain a large number of immature follicles, most of which are destined to degenerate before ovulation [...].
Collapse
Affiliation(s)
- Kenichiro Sakaguchi
- Laboratory of Veterinary Theriogenology, Joint Department of Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, Yanagito 1-1, Gifu 501-1193, Japan;
- Division of Animal Medical Science, Center for One Medicine Innovative Translational Research, Institute for Advanced Study, Gifu University, Yanagito 1-1, Gifu 501-1193, Japan
| |
Collapse
|
17
|
Shirasawa A, Hayashi M, Shono M, Ideta A, Yoshino T, Hayashi K. Efficient derivation of embryonic stem cells and primordial germ cell-like cells in cattle. J Reprod Dev 2024; 70:82-95. [PMID: 38355134 PMCID: PMC11017101 DOI: 10.1262/jrd.2023-087] [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: 10/24/2023] [Accepted: 01/06/2024] [Indexed: 02/16/2024] Open
Abstract
The induction of the germ cell lineage from pluripotent stem cells (in vitro gametogenesis) will help understand the mechanisms underlying germ cell differentiation and provide an alternative source of gametes for reproduction. This technology is especially important for cattle, which are among the most important livestock species for milk and meat production. Here, we developed a new method for robust induction of primordial germ cell-like cells (PGCLCs) from newly established bovine embryonic stem (bES) cells. First, we refined the pluripotent culture conditions for pre-implantation embryos and ES cells. Inhibition of RHO increased the number of epiblast cells in the pre-implantation embryos and dramatically improved the efficiency of ES cell establishment. We then determined suitable culture conditions for PGCLC differentiation using bES cells harboring BLIMP1-tdTomato and TFAP2C-mNeonGreen (BTTN) reporter constructs. After a 24-h culture with bone morphogenetic protein 4 (BMP4), followed by three-dimensional culture with BMP4 and a chemical agonist and WNT signaling chemical antagonist, bES cells became positive for the reporters. A set of primordial germ cells (PGC) marker genes, including PRDM1/BLIMP1, TFAP2C, SOX17, and NANOS3, were expressed in BTTN-positive cells. These bovine PGCLCs (bPGCLCs) were isolated as KIT/CD117-positive and CD44-negative cell populations. We anticipate that this method for the efficient establishment of bES cells and induction of PGCLCs will be useful for stem cell-based reproductive technologies in cattle.
Collapse
Affiliation(s)
- Atsushi Shirasawa
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
- Zen-noh Embryo Transfer Center, Fukuoka 810-0001, Japan
| | - Masafumi Hayashi
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
- Department of Genome Biology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Mayumi Shono
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
- Department of Genome Biology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Atsushi Ideta
- Zen-noh Embryo Transfer Center, Fukuoka 810-0001, Japan
| | - Takashi Yoshino
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Katsuhiko Hayashi
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
- Department of Genome Biology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
- Graduate School of Frontier Biosciences, Osaka University, Osaka 565-0871, Japan
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
18
|
Wu Y, Wang C, Fan X, Ma Y, Liu Z, Ye X, Shen C, Wu C. The impact of induced pluripotent stem cells in animal conservation. Vet Res Commun 2024; 48:649-663. [PMID: 38228922 DOI: 10.1007/s11259-024-10294-3] [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: 11/02/2023] [Accepted: 01/04/2024] [Indexed: 01/18/2024]
Abstract
It is widely acknowledged that we are currently facing a critical tipping point with regards to global extinction, with human activities driving us perilously close to the brink of a devastating sixth mass extinction. As a promising option for safeguarding endangered species, induced pluripotent stem cells (iPSCs) hold great potential to aid in the preservation of threatened animal populations. For endangered species, such as the northern white rhinoceros (Ceratotherium simum cottoni), supply of embryos is often limited. After the death of the last male in 2019, only two females remained in the world. IPSC technology offers novel approaches and techniques for obtaining pluripotent stem cells (PSCs) from rare and endangered animal species. Successful generation of iPSCs circumvents several bottlenecks that impede the development of PSCs, including the challenges associated with establishing embryonic stem cells, limited embryo sources and immune rejection following embryo transfer. To provide more opportunities and room for growth in our work on animal welfare, in this paper we will focus on the progress made with iPSC lines derived from endangered and extinct species, exploring their potential applications and limitations in animal welfare research.
Collapse
Affiliation(s)
- Yurou Wu
- School of Pharmacy/School of Modem Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China
| | - Chengwei Wang
- School of Pharmacy/School of Modem Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China
| | - Xinyun Fan
- School of Pharmacy/School of Modem Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China
| | - Yuxiao Ma
- Department of Biology, New York University, New York, NY, USA
| | - Zibo Liu
- School of Pharmacy/School of Modem Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China
| | - Xun Ye
- School of Pharmacy/School of Modem Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, People's Republic of China
| | - Chongyang Shen
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, People's Republic of China.
| | - Chunjie Wu
- Innovative Institute of Chinese Medicine and Pharmacy/Academy for Interdiscipline, Chengdu Univesity of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, People's Republic of China.
- Sichuan Engineering Research Center for Endangered Medicinal Animals, Chengdu, China.
| |
Collapse
|
19
|
Wilder AP, Steiner CC, Hendricks S, Haller BC, Kim C, Korody ML, Ryder OA. Genetic load and viability of a future restored northern white rhino population. Evol Appl 2024; 17:e13683. [PMID: 38617823 PMCID: PMC11009427 DOI: 10.1111/eva.13683] [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: 11/28/2023] [Revised: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 04/16/2024] Open
Abstract
As biodiversity loss outpaces recovery, conservationists are increasingly turning to novel tools for preventing extinction, including cloning and in vitro gametogenesis of biobanked cells. However, restoration of populations can be hindered by low genetic diversity and deleterious genetic load. The persistence of the northern white rhino (Ceratotherium simum cottoni) now depends on the cryopreserved cells of 12 individuals. These banked genomes have higher genetic diversity than southern white rhinos (C. s. simum), a sister subspecies that successfully recovered from a severe bottleneck, but the potential impact of genetic load is unknown. We estimated how demographic history has shaped genome-wide genetic load in nine northern and 13 southern white rhinos. The bottleneck left southern white rhinos with more fixed and homozygous deleterious alleles and longer runs of homozygosity, whereas northern white rhinos retained more deleterious alleles masked in heterozygosity. To gauge the impact of genetic load on the fitness of a northern white rhino population restored from biobanked cells, we simulated recovery using fitness of southern white rhinos as a benchmark for a viable population. Unlike traditional restoration, cell-derived founders can be reintroduced in subsequent generations to boost lost genetic diversity and relieve inbreeding. In simulations with repeated reintroduction of founders into a restored population, the fitness cost of genetic load remained lower than that borne by southern white rhinos. Without reintroductions, rapid growth of the restored population (>20-30% per generation) would be needed to maintain comparable fitness. Our results suggest that inbreeding depression from genetic load is not necessarily a barrier to recovery of the northern white rhino and demonstrate how restoration from biobanked cells relieves some constraints of conventional restoration from a limited founder pool. Established conservation methods that protect healthy populations will remain paramount, but emerging technologies hold promise to bolster these tools to combat the extinction crisis.
Collapse
Affiliation(s)
- Aryn P. Wilder
- Conservation GeneticsSan Diego Zoo Wildlife AllianceEscondidoCaliforniaUSA
| | - Cynthia C. Steiner
- Conservation GeneticsSan Diego Zoo Wildlife AllianceEscondidoCaliforniaUSA
| | - Sarah Hendricks
- Conservation GeneticsSan Diego Zoo Wildlife AllianceEscondidoCaliforniaUSA
- Institute for Interdisciplinary Data SciencesUniversity of IdahoMoscowIdahoUSA
| | | | - Chang Kim
- University of CaliforniaSanta Cruz Genomics InstituteSanta CruzCaliforniaUSA
- Department of Neurological SurgeryUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Marisa L. Korody
- Conservation GeneticsSan Diego Zoo Wildlife AllianceEscondidoCaliforniaUSA
| | - Oliver A. Ryder
- Conservation GeneticsSan Diego Zoo Wildlife AllianceEscondidoCaliforniaUSA
| |
Collapse
|
20
|
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
|
21
|
Matsuya S, Fujino K, Imai H, Kusakabe KT, Fujii W, Kano K. Establishment of African pygmy mouse induced pluripotent stem cells using defined doxycycline inducible transcription factors. Sci Rep 2024; 14:3204. [PMID: 38331995 PMCID: PMC10853177 DOI: 10.1038/s41598-024-53687-9] [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: 10/14/2023] [Accepted: 02/03/2024] [Indexed: 02/10/2024] Open
Abstract
Mus minutoides is one of the smallest mammals worldwide; however, the regulatory mechanisms underlying its dwarfism have not been examined. Therefore, we aimed to establish M. minutoides induced pluripotent stem cells (iPSCs) using the PiggyBac transposon system for applications in developmental engineering. The established M. minutoides iPSCs were found to express pluripotency markers and could differentiate into neurons. Based on in vitro differentiation analysis, M. minutoides iPSCs formed embryoid bodies expressing marker genes in all three germ layers. Moreover, according to the in vivo analysis, these cells contributed to the formation of teratoma and development of chimeric mice with Mus musculus. Overall, the M. minutoides iPSCs generated in this study possess properties that are comparable to or closely resemble those of naïve pluripotent stem cells (PSCs). These findings suggest these iPSCs have potential utility in various analytical applications, including methods for blastocyst completion.
Collapse
Affiliation(s)
- Sumito Matsuya
- Laboratory of Developmental Biology, Joint Graduate School of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
- Laboratory of Veterinary Anatomy, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Kagoshima, Japan
| | - Kaoru Fujino
- Laboratory of Developmental Biology, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1, Yoshida, Yamaguchi Prefecture, 7538511, Japan
| | - Hiroyuki Imai
- Laboratory of Veterinary Anatomy, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
- Research Institute for Cell Design Medical Science, Yamaguchi University, Yamaguchi, Japan
| | - Ken Takeshi Kusakabe
- Laboratory of Veterinary Anatomy, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Wataru Fujii
- Laboratory of Biomedical Science, Department of Veterinary Medical Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-Ku, Tokyo, 113-8657, Japan.
- Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan.
| | - Kiyoshi Kano
- Laboratory of Developmental Biology, Joint Graduate School of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan.
- Laboratory of Developmental Biology, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1, Yoshida, Yamaguchi Prefecture, 7538511, Japan.
- Research Institute for Cell Design Medical Science, Yamaguchi University, Yamaguchi, Japan.
| |
Collapse
|
22
|
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
|
23
|
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
|
24
|
Irie N, Kobayashi T, Azim Surani M. Human Primordial Germ Cell-Like Cell Induction from Pluripotent Stem Cells by SOX17 and PRDM1 Expression. Methods Mol Biol 2024; 2770:87-97. [PMID: 38351448 DOI: 10.1007/978-1-0716-3698-5_7] [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
Human primordial germ cell (PGC) development initiates about 2 weeks after fertilization during embryogenesis. Unique molecular events follow, including epigenetic resetting, to establish functional gametes (egg and sperm). Due to the inaccessibility of human embryos, it is essential to have an amenable experimental platform to investigate the mechanisms and potential dysfunctions of the events. We previously established a PGC-like cell (PGCLC) differentiation method using human pluripotent stem cells (PSCs) via induction of precursor cells followed by stimulation with a cytokine cocktail including BMP. We also revealed that the expression of PGC specifiers, SOX17 and PRDM1, can robustly induce PGCLCs from PSCs without the cytokines. The balance of SOX17 and PRDM1 is critical for germ cell fate since the two factors also regulate endoderm differentiation. Here we describe a detailed procedure for PGCLC differentiation with the balanced induction of SOX17 and PRDM1. The protocol can be used for PGC induction in other mammalian species exhibiting PGCs with SOX17 expression. Together, these studies will advance the understanding of germ cell biology and its applications in reproductive technology and medicine.
Collapse
Affiliation(s)
- Naoko Irie
- Wellcome Trust/Cancer Research U.K. Gurdon Institute, Henry Wellcome Building of Cancer and Developmental Biology, Cambridge, UK.
- Metabolic Systems Laboratory, Live Imaging Center, Central Institute for Experimental Animals, Kawasaki-ku, Kanagawa, Japan.
| | - Toshihiro Kobayashi
- Division of Mammalian Embryology, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
- Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
| | - M Azim Surani
- Wellcome Trust/Cancer Research U.K. Gurdon Institute, Henry Wellcome Building of Cancer and Developmental Biology, Cambridge, UK.
- Physiology, Development and Neuroscience Department, University of Cambridge, Cambridge, UK.
| |
Collapse
|
25
|
Wang X, Zhu J, Wang H, Deng W, Jiao S, Wang Y, He M, Zhang F, Liu T, Hao Y, Ye D, Sun Y. Induced formation of primordial germ cells from zebrafish blastomeres by germplasm factors. Nat Commun 2023; 14:7918. [PMID: 38097571 PMCID: PMC10721796 DOI: 10.1038/s41467-023-43587-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 11/14/2023] [Indexed: 12/17/2023] Open
Abstract
The combination of genome editing and primordial germ cell (PGC) transplantation has enormous significance in the study of developmental biology and genetic breeding, despite its low efficiency due to limited number of donor PGCs. Here, we employ a combination of germplasm factors to convert blastoderm cells into induced PGCs (iPGCs) in zebrafish and obtain functional gametes either through iPGC transplantation or via the single blastomere overexpression of germplasm factors. Zebrafish-derived germplasm factors convert blastula cells of Gobiocypris rarus into iPGCs, and Gobiocypris rarus spermatozoa can be produced by iPGC-transplanted zebrafish. Moreover, the combination of genome knock-in and iPGC transplantation perfectly resolves the contradiction between high knock-in efficiency and early lethality during embryonic stages and greatly improves the efficiency of genome knock-in. Together, we present an efficient method for generating PGCs in a teleost, a technique that will have a strong impact in basic research and aquaculture.
Collapse
Affiliation(s)
- Xiaosi Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Junwen Zhu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Houpeng Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Wenqi Deng
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shengbo Jiao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yaqing Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Mudan He
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Fenghua Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tao Liu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yongkang Hao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ding Ye
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Yonghua Sun
- State Key Laboratory of Freshwater Ecology and Biotechnology, Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China.
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
- Hubei Hongshan Laboratory, Wuhan, 430070, China.
| |
Collapse
|
26
|
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
|
27
|
Hildebrandt TB, Holtze S, Colleoni S, Hermes R, Stejskal J, Lekolool I, Ndeereh D, Omondi P, Kariuki L, Mijele D, Mutisya S, Ngulu S, Diecke S, Hayashi K, Lazzari G, de Mori B, Biasetti P, Quaggio A, Galli C, Goeritz F. In vitro fertilization program in white rhinoceros. Reproduction 2023; 166:383-399. [PMID: 37877686 PMCID: PMC10620463 DOI: 10.1530/rep-23-0087] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 09/19/2023] [Indexed: 09/22/2023]
Abstract
In brief To save endangered rhinoceros species, assisted reproductive technologies are warranted. We here report in vitro blastocyst generation of the Near-Threatened Southern white rhinoceros and, for the first time, also of the technically Extinct Northern white rhinoceros. Abstract The Anthropocene is marked by a dramatic biodiversity decline, particularly affecting the family Rhinocerotidae. Three of five extant species are listed as Critically Endangered (Sumatran, Javan, black rhinoceros), one as Vulnerable (Indian rhinoceros), and only one white rhino (WR) subspecies, the Southern white rhinoceros (SWR), after more than a century of successful protection is currently classified as Near Threatened by the IUCN, while numbers again are declining. Conversely, in 2008, the SWR's northern counterpart and second WR subspecies, the Northern white rhinoceros (NWR), was declared extinct in the wild. Safeguarding these vanishing keystone species urgently requires new reproductive strategies. We here assess one such strategy, the novel in vitro fertilization program in SWR and - for the first-time NWR - regarding health effects, donor-related, and procedural factors. Over the past 8 years, we performed 65 procedures in 22 white rhinoceros females (20 SWR and 2 NWR) comprising hormonal ovarian stimulation, ovum pick-up (OPU), in vitro oocyte maturation, fertilization, embryo culture, and blastocyst cryopreservation, at an efficiency of 1.0 ± 1.3 blastocysts per OPU, generating 22 NWR, 19 SWR and 10 SWR/NWR hybrid blastocysts for the future generation of live offspring.
Collapse
Affiliation(s)
- Thomas Bernd Hildebrandt
- Leibniz Institute for Zoo and Wildlife Research (IZW) in the Forschungsverbund Berlin eV, Reproduction Management, Alfred-Kowalke-Straße, Berlin, Germany
- Freie Universitat Berlin, Veterinary Medicine, Berlin, Germany
| | - Susanne Holtze
- Leibniz Institute for Zoo and Wildlife Research (IZW) in the Forschungsverbund Berlin eV, Reproduction Management, Alfred-Kowalke-Straße, Berlin, Germany
| | - Silvia Colleoni
- AVANTEA, Laboratorio di Tecnologie della Riproduzione, Lombardy, Cremona, Italy
| | - Robert Hermes
- Leibniz Institute for Zoo and Wildlife Research (IZW) in the Forschungsverbund Berlin eV, Reproduction Management, Alfred-Kowalke-Straße, Berlin, Germany
| | - Jan Stejskal
- ZOO Dvůr Králové, Communication and International Projects, Štefánikova, Dvůr Králové nad Labem, Czech Republic
| | - Isaac Lekolool
- Kenya Wildlife Service, Veterinary and Capture Services, Nairobi, Kenya
| | - David Ndeereh
- Wildlife Training and Research Institute, Nakuru County, Naivasha, Kenya
| | - Patrick Omondi
- Kenya Wildlife Service, Veterinary and Capture Services, Nairobi, Kenya
| | - Linus Kariuki
- Kenya Wildlife Service, Veterinary and Capture Services, Nairobi, Kenya
| | - Domnic Mijele
- Kenya Wildlife Service, Veterinary and Capture Services, Nairobi, Kenya
| | - Samuel Mutisya
- Ol Pejeta Conservancy, Conservation Laikipia, Nanyuki, Kenya
| | - Stephen Ngulu
- Ol Pejeta Conservancy, Conservation Laikipia, Nanyuki, Kenya
| | - Sebastian Diecke
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Technology Platform Pluripotent Stem Cells, Berlin, Germany
| | - Katsuhiko Hayashi
- Department of Stem Cell Biology and Medicine, Kyushu University, Maidashi, Higashiku, Fukuoka, Japan
- Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Giovanna Lazzari
- AVANTEA, Laboratorio di Tecnologie della Riproduzione, Lombardy, Cremona, Italy
| | - Barbara de Mori
- Department of Comparative Biomedicine and Food Science, Università degli Studi di Padova, Italy
- Universita degli Studi di Padova, Ethics Laboratory for Veterinary Medicine, Conservation, and Animal Welfare, Veneto, Padova, Italy
| | - Pierfrancesco Biasetti
- Leibniz Institute for Zoo and Wildlife Research (IZW) in the Forschungsverbund Berlin eV, Reproduction Management, Alfred-Kowalke-Straße, Berlin, Germany
- Universita degli Studi di Padova, Ethics Laboratory for Veterinary Medicine, Conservation, and Animal Welfare, Veneto, Padova, Italy
| | - Alessandra Quaggio
- Leibniz Institute for Zoo and Wildlife Research (IZW) in the Forschungsverbund Berlin eV, Reproduction Management, Alfred-Kowalke-Straße, Berlin, Germany
| | - Cesare Galli
- AVANTEA, Laboratorio di Tecnologie della Riproduzione, Lombardy, Cremona, Italy
- Fondazione Avantea, Riproduzione Cremona, Lombardy, Cremona, Italy
| | - Frank Goeritz
- Leibniz Institute for Zoo and Wildlife Research (IZW) in the Forschungsverbund Berlin eV, Reproduction Management, Alfred-Kowalke-Straße, Berlin, Germany
| |
Collapse
|
28
|
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
|
29
|
Liu YL, Chen JS, An JH, Cai ZG, Lan JC, Li Y, Kong XW, Zhang MY, Hou R, Wang DH. Characteristics of mesenchymal stem cells and their exosomes derived from giant panda (Ailuropoda melanoleuca) endometrium. In Vitro Cell Dev Biol Anim 2023; 59:550-563. [PMID: 37639049 DOI: 10.1007/s11626-023-00802-1] [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: 05/26/2023] [Accepted: 07/27/2023] [Indexed: 08/29/2023]
Abstract
Conservation of genetic resources is an important way to protect endangered species. At present, mesenchymal stem cells (MSCs) have been isolated from the bone marrow and umbilical cords of giant pandas. However, the types and quantities of preserved cell resources were rare and limited, and none of MSCs was derived from female reproductive organs. Here, we first isolated MSCs from the endometrium of giant panda. These cells showed fibroblast morphology and expressed Sox2, Klf4, Thy1, CD73, CD105, CD44, CD49f, and CD105. Endometrium mesenchymal stem cells (eMSCs) of giant panda could induce differentiation into three germ layers in vitro. RNA-seq analysis showed that 833 genes were upregulated and 716 genes were downregulated in eMSCs compared with skin fibroblast cells. The results of GO and the KEGG analysis of differentially expressed genes (DEGs) were mainly focused on transporter activity, signal transducer activity, pathways regulating pluripotency of stem cells, MAPK signaling pathway, and PI3K-Akt signaling pathway. The genes PLCG2, FRK, JAK3, LYN, PIK3CB, JAK2, CBLB, and MET were identified as hub genes by PPI network analysis. In addition, the exosomes of eMSCs were also isolated and identified. The average diameter of exosomes was 74.26 ± 13.75 nm and highly expressed TSG101 and CD9 but did not express CALNEXIN. A total of 277 miRNAs were detected in the exosomes; the highest expression of miRNA was the has-miR-21-5p. A total of 14461 target genes of the whole miRNAs were predicted and proceeded with functional analysis. In conclusion, we successfully isolated and characterized the giant panda eMSCs and their exosomes, and analyzed their functions through bioinformatics techniques. It not only enriched the conservation types of giant panda cell resources and promoted the protection of genetic diversity, but also laid a foundation for the application of eMSCs and exosomes in the disease treatment of giant pandas.
Collapse
Affiliation(s)
- Yu-Liang Liu
- Chengdu Research Base of Giant Panda Breeding, Sichuan Province, Chengdu, 610081, China
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Sichuan Province, Chengdu, 610081, China
- Sichuan Academy of Giant Panda, Sichuan Province, Chengdu, 610081, China
| | - Jia-Song Chen
- Chengdu Research Base of Giant Panda Breeding, Sichuan Province, Chengdu, 610081, China
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Sichuan Province, Chengdu, 610081, China
- Sichuan Academy of Giant Panda, Sichuan Province, Chengdu, 610081, China
| | - Jun-Hui An
- Chengdu Research Base of Giant Panda Breeding, Sichuan Province, Chengdu, 610081, China
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Sichuan Province, Chengdu, 610081, China
- Sichuan Academy of Giant Panda, Sichuan Province, Chengdu, 610081, China
| | - Zhi-Gang Cai
- Chengdu Research Base of Giant Panda Breeding, Sichuan Province, Chengdu, 610081, China
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Sichuan Province, Chengdu, 610081, China
- Sichuan Academy of Giant Panda, Sichuan Province, Chengdu, 610081, China
| | - Jing-Chao Lan
- Chengdu Research Base of Giant Panda Breeding, Sichuan Province, Chengdu, 610081, China
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Sichuan Province, Chengdu, 610081, China
- Sichuan Academy of Giant Panda, Sichuan Province, Chengdu, 610081, China
| | - Yuan Li
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Sichuan Province, Chengdu, 610081, China
| | - Xiang-Wei Kong
- Chengdu Research Base of Giant Panda Breeding, Sichuan Province, Chengdu, 610081, China
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Sichuan Province, Chengdu, 610081, China
- Sichuan Academy of Giant Panda, Sichuan Province, Chengdu, 610081, China
| | - Ming-Yue Zhang
- Chengdu Research Base of Giant Panda Breeding, Sichuan Province, Chengdu, 610081, China
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Sichuan Province, Chengdu, 610081, China
- Sichuan Academy of Giant Panda, Sichuan Province, Chengdu, 610081, China
| | - Rong Hou
- Chengdu Research Base of Giant Panda Breeding, Sichuan Province, Chengdu, 610081, China
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Sichuan Province, Chengdu, 610081, China
- Sichuan Academy of Giant Panda, Sichuan Province, Chengdu, 610081, China
| | - Dong-Hui Wang
- Chengdu Research Base of Giant Panda Breeding, Sichuan Province, Chengdu, 610081, China.
- Sichuan Key Laboratory of Conservation Biology for Endangered Wildlife, Sichuan Province, Chengdu, 610081, China.
- Sichuan Academy of Giant Panda, Sichuan Province, Chengdu, 610081, China.
| |
Collapse
|
30
|
Lázaro J, Costanzo M, Sanaki-Matsumiya M, Girardot C, Hayashi M, Hayashi K, Diecke S, Hildebrandt TB, Lazzari G, Wu J, Petkov S, Behr R, Trivedi V, Matsuda M, Ebisuya M. A stem cell zoo uncovers intracellular scaling of developmental tempo across mammals. Cell Stem Cell 2023; 30:938-949.e7. [PMID: 37343565 PMCID: PMC10321541 DOI: 10.1016/j.stem.2023.05.014] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 04/03/2023] [Accepted: 05/25/2023] [Indexed: 06/23/2023]
Abstract
Differential speeds in biochemical reactions have been proposed to be responsible for the differences in developmental tempo between mice and humans. However, the underlying mechanism controlling the species-specific kinetics remains to be determined. Using in vitro differentiation of pluripotent stem cells, we recapitulated the segmentation clocks of diverse mammalian species varying in body weight and taxa: marmoset, rabbit, cattle, and rhinoceros. Together with mouse and human, the segmentation clock periods of the six species did not scale with the animal body weight, but with the embryogenesis length. The biochemical kinetics of the core clock gene HES7 displayed clear scaling with the species-specific segmentation clock period. However, the cellular metabolic rates did not show an evident correlation. Instead, genes involving biochemical reactions showed an expression pattern that scales with the segmentation clock period. Altogether, our stem cell zoo uncovered general scaling laws governing species-specific developmental tempo.
Collapse
Affiliation(s)
- Jorge Lázaro
- European Molecular Biology Laboratory (EMBL) Barcelona, Dr. Aiguader 88, 08003 Barcelona, Spain; Collaboration for joint PhD degree between EMBL and Heidelberg University, Faculty of Biosciences, Heidelberg, Germany
| | - Maria Costanzo
- European Molecular Biology Laboratory (EMBL) Barcelona, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Marina Sanaki-Matsumiya
- European Molecular Biology Laboratory (EMBL) Barcelona, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Charles Girardot
- European Molecular Biology Laboratory, Genome Biology Unit, 69117 Heidelberg, Germany
| | - Masafumi Hayashi
- Department of Genome Biology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Katsuhiko Hayashi
- Department of Genome Biology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Sebastian Diecke
- Technology Platform Pluripotent Stem Cells, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | | | | | - Jun Wu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9148, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-9148, USA
| | - Stoyan Petkov
- Platform Degenerative Diseases, German Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany; German Center for Cardiovascular Research (DZHK), Partner site Göttingen, Göttingen, Germany
| | - Rüdiger Behr
- Platform Degenerative Diseases, German Primate Center - Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany; German Center for Cardiovascular Research (DZHK), Partner site Göttingen, Göttingen, Germany
| | - Vikas Trivedi
- European Molecular Biology Laboratory (EMBL) Barcelona, Dr. Aiguader 88, 08003 Barcelona, Spain.
| | - Mitsuhiro Matsuda
- European Molecular Biology Laboratory (EMBL) Barcelona, Dr. Aiguader 88, 08003 Barcelona, Spain.
| | - Miki Ebisuya
- European Molecular Biology Laboratory (EMBL) Barcelona, Dr. Aiguader 88, 08003 Barcelona, Spain; Cluster of Excellence Physics of Life, TU Dresden, Dresden, Germany.
| |
Collapse
|
31
|
Wesevich VG, Arkfeld C, Seifer DB. In Vitro Gametogenesis in Oncofertility: A Review of Its Potential Use and Present-Day Challenges in Moving toward Fertility Preservation and Restoration. J Clin Med 2023; 12:3305. [PMID: 37176745 PMCID: PMC10179531 DOI: 10.3390/jcm12093305] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/12/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
Current fertility preservation options are limited for cancer survivor patients who wish to have their own biological children. Human in vitro gametogenesis (IVG) has the hypothetical ability to offer a unique solution to individuals receiving treatment for cancer which subsequently shortens their reproductive lifespan. Through a simple skin punch biopsy, a patient's fertility could be restored via reprogramming of dermal fibroblast cells to induced pluripotent stem cells, then from primordial germ cell-like cells into viable oocytes and spermatocytes which could be used for embryogenesis. Induced pluripotent stem cells could also be used to form in vitro environments, similar to the ovary or testes, necessary for the maturation of oogonia. This would allow for the entire creation of embryos outside the body, ex vivo. While this area in stem cell biology research offers the potential to revolutionize reproduction as we know it, there are many critical barriers, both scientific and ethical, that need to be overcome to one day see this technology utilized clinically.
Collapse
Affiliation(s)
- Victoria G Wesevich
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510, USA
| | - Christopher Arkfeld
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale New Haven Hospital, New Haven, CT 06510, USA
| | - David B Seifer
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
32
|
Strange A, Alberio R. Review: A barnyard in the lab: prospect of generating animal germ cells for breeding and conservation. Animal 2023; 17 Suppl 1:100753. [PMID: 37567650 DOI: 10.1016/j.animal.2023.100753] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 02/14/2023] [Accepted: 02/20/2023] [Indexed: 08/13/2023] Open
Abstract
In vitro gametogenesis (IVG) offers broad opportunities for gaining detailed new mechanistic knowledge of germ cell biology that will enable progress in the understanding of human infertility, as well as for applications in the conservation of endangered species and for accelerating genetic selection of livestock. The realisation of this potential depends on overcoming key technical challenges and of gaining more detailed knowledge of the ontogeny and developmental programme in different species. Important differences in the molecular mechanisms of germ cell determination and epigenetic reprogramming between mice and other animals have been elucidated in recent years. These must be carefully considered when developing IVG protocols, as cellular kinetics in mice may not accurately reflect mechanisms in other mammals. Similarly, diverse stem cell models with potential for germ cell differentiation may reflect alternative routes to successful IVG. In conclusion, the fidelity of the developmental programme recapitulated during IVG must be assessed against reference information from each species to ensure the production of healthy animals using these methods, as well as for developing genuine models of gametogenesis.
Collapse
Affiliation(s)
- A Strange
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, LE12 5RD, UK
| | - R Alberio
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, LE12 5RD, UK.
| |
Collapse
|
33
|
Menzorov AG. Pluripotent Stem Cells of Order Carnivora: Technical Perspective. Int J Mol Sci 2023; 24:ijms24043905. [PMID: 36835318 PMCID: PMC9963171 DOI: 10.3390/ijms24043905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/08/2023] [Accepted: 02/12/2023] [Indexed: 02/17/2023] Open
Abstract
Human and mouse induced pluripotent stem cells (PSCs) are widely used for studying early embryonic development and for modeling of human diseases. Derivation and studying of PSCs from model organisms beyond commonly used mice and rats may provide new insights into the modeling and treating human diseases. The order Carnivora representatives possess unique features and are already used for modeling human-related traits. This review focuses on the technical aspects of derivation of the Carnivora species PSCs as well as their characterization. Current data on dog, feline, ferret, and American mink PSCs are summarized.
Collapse
Affiliation(s)
- Aleksei G. Menzorov
- Sector of Cell Collections, Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia;
- Natural Sciences Department, Novosibirsk State University, 630090 Novosibirsk, Russia
| |
Collapse
|