1
|
Scalf SM, Wu Q, Guo S. Molecular basis of cell fate plasticity - insights from the privileged cells. Curr Opin Genet Dev 2025; 93:102354. [PMID: 40327951 DOI: 10.1016/j.gde.2025.102354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 05/08/2025]
Abstract
In the post-Yamanaka era, the rolling balls on Waddington's hilly landscape not only roll downward, but also go upward or sideways. This new-found mobility implies that the tantalizing somatic cell plasticity fueling regeneration, once only known to planarians and newts, might be sparking in the cells of mice and humans, if only we knew how to fully unlock it. The hope for ultimate regeneration was made even more tangible by the observations that partial reprogramming by the Yamanaka factors reverses many hallmarks of aging [76], even though the underlying mechanism remains unclear. We intend to revisit the milestones in the evolving understanding of cell fate plasticity and glean molecular insights from an unusual somatic cell state, the privileged cell state that reprograms in a manner defying the stochastic model. We synthesize our view of the molecular underpinning of cell fate plasticity, from which we speculate how to harness it for regeneration and rejuvenation. We propose that senescence, aging and malignancy represent distinct cell states with definable biochemical and biophysical parameters.
Collapse
Affiliation(s)
- Stephen Maxwell Scalf
- Department of Cell Biology, Yale University, Yale Stem Cell Center, Yale University, United States
| | - Qiao Wu
- Department of Cell Biology, Yale University, Yale Stem Cell Center, Yale University, United States
| | - Shangqin Guo
- Department of Cell Biology, Yale University, Yale Stem Cell Center, Yale University, United States.
| |
Collapse
|
2
|
Su R, Ding F, Sorgog M, Li L, Bai S, Ritu X, Nasenochir N, Fan R, Herrid M. Optimization of handmade cloning enhances pregnancy rates and live calf production in cattle. Sci Rep 2025; 15:15032. [PMID: 40301396 PMCID: PMC12041346 DOI: 10.1038/s41598-025-96511-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 03/28/2025] [Indexed: 05/01/2025] Open
Abstract
Handmade cloning (HMC) presents several advantages over conventional cloning, including higher throughput, cost-efficiency, and operational simplicity. However, a comprehensive analysis comparing embryo production rates and pregnancy outcomes in cattle has yet to be conducted. This study reveals that cytoplasts produced using the micropipette method exhibited higher quality than those created with a microblade, leading to improved cleavage and blastocyst rates. The fusion of one or double cytoplast did not significantly affect the developmental potential of reconstructed embryos, although blastocysts from single cytoplasts contained slightly fewer cells. Notably, HMC demonstrated higher pregnancy rates and live calf production efficiency compared to conventional cloning. Specifically, for 41 vitrified embryos from conventional cloning, the one-month post-transfer pregnancy rate was 41.4%, resulting in 6 healthy calves (14.6%). In contrast, HMC produced one-month pregnancy rates of 71.4% for fresh and 60.0% for vitrified embryos, yielding 6 (28.5%) and 4 (26.7%) healthy calves, respectively. The optimization of HMC emphasizes the micropipette method's potential for cattle cloning applications, including genomic selection and gene editing. Additionally, novel insights into the incompatibility issues between nuclear DNA and mitochondrial DNA (mtDNA) in cloned embryos are discussed.
Collapse
Affiliation(s)
- Rina Su
- Grassland & Cattle Investment Co., Ltd., Hohhot, 010000, Inner Mongolia, China
| | - Fangrong Ding
- College of Biological Sciences, Chinese Agricultural University, Beijing, 100000, China
| | - Moqir Sorgog
- Grassland & Cattle Investment Co., Ltd., Hohhot, 010000, Inner Mongolia, China
| | - Ling Li
- College of Biological Sciences, Chinese Agricultural University, Beijing, 100000, China
| | - Suyilatu Bai
- Grassland & Cattle Investment Co., Ltd., Hohhot, 010000, Inner Mongolia, China
| | - Xiji Ritu
- Grassland & Cattle Investment Co., Ltd., Hohhot, 010000, Inner Mongolia, China
| | - Narenhua Nasenochir
- College of Animal Science, Inner Mongolia Agriculture University, Hohhot, 010000, Inner Mongolia, China
| | - Ruiwen Fan
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, China
| | - Muren Herrid
- Grassland & Cattle Investment Co., Ltd., Hohhot, 010000, Inner Mongolia, China.
- Internaional Livestock Research Centre, Gold Coast, Queensland, 4211, Australia.
| |
Collapse
|
3
|
Dunn IC. The Gordon Memorial Lecture: genotype, phenotype, selection and more: improving the skeletal health of laying hens. Br Poult Sci 2025; 66:139-146. [PMID: 40052830 PMCID: PMC11974920 DOI: 10.1080/00071668.2025.2460054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 01/16/2025] [Indexed: 04/03/2025]
Abstract
1. This review is a comprehensive exploration of the author's work in improving skeletal health in laying hens, focusing on the insights from genetics on nutritional, and environmental factors. It discusses the importance of the large number of disciplines that have contributed to the efforts to tackle bone quality in laying hens, particularly the keel bone.2. The transition from cages to non-cage environments has increased keel bone damage, despite improving overall skeletal health. It is a welfare paradox that improving the hen's environment has often been accompanied by greater skeletal damage.3. The role of genetics has been important in understanding and addressing bone health issues and will be a major factor in their improvement. This includes the identification of specific genes, like cystathionine-β-synthase, which has led to nutritional interventions using betaine supplementation to improve bone quality by targeting the one carbon pathway.4. The role of the timing of puberty and its genetic control is an additional factor in bone health, and new methods of measuring bone density in live birds are now important to monitor potential issues and deliver genetic solutions.5. The review emphasises a multi-faceted approach, combining genetics, nutrition, rearing practices, and housing design is required in order to improve skeletal health and enhance the welfare and sustainable performance in laying hens.
Collapse
Affiliation(s)
- I. C. Dunn
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
4
|
de Macedo MP, Glanzner WG, Gutierrez K, Currin L, Rissi VB, Baldassarre H, McGraw S, Bordignon V. Heterologous expression of bovine histone H1foo into porcine fibroblasts alters the transcriptome profile but not embryo development following nuclear transfer. J Assist Reprod Genet 2025; 42:1109-1120. [PMID: 40025368 PMCID: PMC12055672 DOI: 10.1007/s10815-025-03437-1] [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: 08/14/2024] [Accepted: 02/18/2025] [Indexed: 03/04/2025] Open
Abstract
PURPOSE Somatic cell nuclear transfer (SCNT) is a valuable tool for investigating reprogramming mechanisms and creating animal clones for applications in production, conservation, companionship, and biomedical research. However, SCNT efficiency remains low. Expression of nuclear proteins associated with an undifferentiated chromatin state, such as the oocyte-specific variant of the linker histone H1 (H1foo), represents a strategy for improving reprogramming outcomes, but this approach has not been tested in the context of SCNT. METHODS Bovine H1foo (bH1foo) was transfected into porcine fibroblasts via electroporation for expression until SCNT. The transcriptomic profile of these cells was analyzed, and their potential as donor cells for SCNT was evaluated 48 h post-electroporation. RESULTS Strong nuclear localization of bH1foo persisted for 48 h post-electroporation. A total of 447 genes were differentially expressed, and lower levels of H3K4me3 and H3K27me3 were detected in bH1foo-expressing cells, indicating changes in chromatin remodeling and function. Embryo development and total cell number per blastocyst were similar between SCNT embryos produced with control and bH1foo-expressing cells. mRNA levels of genes involved in embryonic genome activation were comparable between embryos derived from control and bH1foo-expressing cells on days 3 and 4 of development, suggesting that bH1foo did not disrupt this critical process. CONCLUSIONS The heterologous expression of bovine H1foo altered the chromatin function of porcine fibroblasts without impairing development to the blastocyst stage following SCNT. These results highlight the potential of expressing nuclear proteins as a strategy to enhance cell reprogramming and cloning efficiency, including interspecies cloning applications.
Collapse
Affiliation(s)
- Mariana Priotto de Macedo
- Department of Animal Science, Mcgill University, 21111 Lakeshore Road, Sainte-Anne-de-Bellevue, Montreal, QC, H9X 3V9, Canada
| | - Werner Giehl Glanzner
- Department of Animal Science, Mcgill University, 21111 Lakeshore Road, Sainte-Anne-de-Bellevue, Montreal, QC, H9X 3V9, Canada
| | - Karina Gutierrez
- Department of Animal Science, Mcgill University, 21111 Lakeshore Road, Sainte-Anne-de-Bellevue, Montreal, QC, H9X 3V9, Canada
| | - Luke Currin
- Department of Animal Science, Mcgill University, 21111 Lakeshore Road, Sainte-Anne-de-Bellevue, Montreal, QC, H9X 3V9, Canada
| | - Vitor Braga Rissi
- Department of Agriculture, Biodiversity and Forests, Federal University of Santa Catarina, Curitibanos, SC, Brazil
| | - Hernan Baldassarre
- Department of Animal Science, Mcgill University, 21111 Lakeshore Road, Sainte-Anne-de-Bellevue, Montreal, QC, H9X 3V9, Canada
| | - Serge McGraw
- Azrieli Research Centre of Sainte-Justine University Hospital, Montreal, QC, Canada
- Department of Obstetrics and Gynecology, University of Montreal, Montreal, QC, Canada
| | - Vilceu Bordignon
- Department of Animal Science, Mcgill University, 21111 Lakeshore Road, Sainte-Anne-de-Bellevue, Montreal, QC, H9X 3V9, Canada.
| |
Collapse
|
5
|
Qin Y. Nearly a Century of Nuclear Transfer Research: Milestones, Applications, and Challenges. Cell Reprogram 2025; 27:56-74. [PMID: 40064535 DOI: 10.1089/cell.2024.0089] [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: 04/05/2025] Open
Abstract
From the first cloning of animals-salamanders-to the cloning of primates-monkeys-nuclear transfer research has spanned an extensive 96-year history. Over the course of nearly a century, it has addressed fundamental scientific questions and found applications across a wide range of practical fields. This review provides a comprehensive overview of the key milestones in its development, its practical applications, and the challenges it continues to face.
Collapse
Affiliation(s)
- Yiren Qin
- Black Family Stem Cell Institute, Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
6
|
Novak BJ, Brand S, Phelan R, Plichta S, Ryder OA, Wiese RJ. Towards Practical Conservation Cloning: Understanding the Dichotomy Between the Histories of Commercial and Conservation Cloning. Animals (Basel) 2025; 15:989. [PMID: 40218382 PMCID: PMC11988126 DOI: 10.3390/ani15070989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Over 40 years ago, scientists imagined ways cloning could aid conservation of threatened taxa. The cloning of Dolly the sheep from adult somatic cells in 1996 was the breakthrough that finally enabled the conservation potential of the technology. Until the 2020s, conservation cloning research efforts yielded no management applications, leading many to believe cloning is not yet an effective conservation tool. In strong contrast, domestic taxa are cloned routinely for scientific and commercial purposes. In this review, we sought to understand the reasons for these divergent trends. We scoured peer-reviewed and gray literature and sent direct inquiries to scientists to analyze a more comprehensive history of the field than was analyzed in previous reviews. While most previous reviewers concluded that a lack of reproductive knowledge of wildlife species has hindered advances for wider conservation applications, we found that resource limitations (e.g., numbers of surrogates, sustainable funding) and widely held misconceptions about cloning are significant contributors to the stagnation of the field. Recent successes in cloning programs for the endangered black-footed ferret (Mustela nigripes) and Przewalski's horse (Equus przewalskii), the world's first true applied-conservation cloning efforts, are demonstrating that cloning can be used for significant conservation impact in the present. When viewed alongside the long history of cloning achievements, these programs emphasize the value of investing in the science and resources needed to meaningfully integrate cloning into conservation management, especially for species with limited genetic diversity that rely on the maintenance of small populations for many generations while conservationists work to restore habitat and mitigate threats in the wild.
Collapse
Affiliation(s)
- Ben J. Novak
- Revive & Restore, 1505 Bridgeway #203, Sausalito, CA 94965, USA
| | - Stewart Brand
- Revive & Restore, 1505 Bridgeway #203, Sausalito, CA 94965, USA
| | - Ryan Phelan
- Revive & Restore, 1505 Bridgeway #203, Sausalito, CA 94965, USA
| | - Sasha Plichta
- Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Oliver A. Ryder
- Beckman Center for Conservation Research, San Diego Zoo Wildlife Alliance, Escondido, CA 92027, USA
| | - Robert J. Wiese
- North Carolina Museum of Natural Sciences, Raleigh, NC 27601, USA;
| |
Collapse
|
7
|
Saadeldin IM, Ehab S, Alshammari MEF, Abdelazim AM, Assiri AM. The Mammalian Oocyte: A Central Hub for Cellular Reprogramming and Stemness. Stem Cells Cloning 2025; 18:15-34. [PMID: 39991743 PMCID: PMC11846613 DOI: 10.2147/sccaa.s513982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 02/13/2025] [Indexed: 02/25/2025] Open
Abstract
The mammalian oocyte is pivotal in reproductive biology, acting as a central hub for cellular reprogramming and stemness. It uniquely contributes half of the zygotic nuclear genome and the entirety of the mitochondrial genome, ensuring individual development and health. Oocyte-mediated reprogramming, exemplified by nuclear transfer, resets somatic cell identity to achieve pluripotency and has transformative potential in regenerative medicine. This process is critical for understanding cellular differentiation, improving assisted reproductive technologies, and advancing cloning and stem cell research. During fertilization, the maternal-zygotic transition shifts developmental control from maternal factors to zygotic genome activation, establishing totipotency. Oocytes also harbor reprogramming factors that guide nuclear remodeling, epigenetic modifications, and metabolic reprogramming, enabling early embryogenesis. Structures like mitochondria, lipid droplets, and cytoplasmic lattices contribute to energy production, molecular regulation, and cellular organization. Recent insights into oocyte components, such as ooplasmic nanovesicles and endolysosomal vesicular assemblies (ELVAS), highlight their roles in maintaining cellular homeostasis, protein synthesis, and reprogramming efficiency. By unraveling the reprogramming mechanisms inherent in oocytes, we advance our understanding of cloning, cell differentiation, and stem cell therapy, highlighting their valuable significance in developmental biology and regenerative medicine.
Collapse
Affiliation(s)
- Islam M Saadeldin
- Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, 11211, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, 11533, Saudi Arabia
| | - Seif Ehab
- Department of Zoology, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | | | - Aaser M Abdelazim
- Department of Medical Laboratories Sciences, College of Applied Medical Sciences, University of Bisha, Bisha, 67714, Saudi Arabia
| | - Abdullah M Assiri
- Comparative Medicine Department, King Faisal Specialist Hospital and Research Centre, Riyadh, 11211, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, 11533, Saudi Arabia
| |
Collapse
|
8
|
Wakayama S, Wakayama T. Can Humanity Thrive Beyond the Galaxy? J Reprod Dev 2025; 71:10-16. [PMID: 39756865 PMCID: PMC11808306 DOI: 10.1262/jrd.2024-099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 12/19/2024] [Indexed: 01/07/2025] Open
Abstract
In the future, human beings will surely expand into space. But given its unique risks, will humanity thrive in space environments? For example, when humans begin living and reproducing in space habitats or on other planets in the solar system, are there risks that future generations may suffer from adverse mutations induced by space radiation, or that embryos and fetuses will develop abnormally in gravitational environments that differ from that of Earth? Moreover, human expansion to other stellar systems requires that for each breed of animal, thousands of individuals must be transported to destination planets to prevent populations from experiencing inbreeding-related degeneration. In even more distant future, when humans have spread throughout the galaxy, all genetic resources on Earth, the planet where humans originated, must be permanently and safely stored- but is this even possible? Such issues with future space colonization may not be an urgent research priority, but research and technological development accompanying advancements in spaceflight will excite many people and contribute to technological improvements that can improve living standards in the present day (e.g., more effective treatments for infertility, etc.). This review will therefore focus primarily on issues related to mammalian reproduction in space environments.
Collapse
Affiliation(s)
- Sayaka Wakayama
- Advanced Biotechnology Center, University of Yamanashi, Yamanashi 400-8510, Japan
| | - Teruhiko Wakayama
- Advanced Biotechnology Center, University of Yamanashi, Yamanashi 400-8510, Japan
| |
Collapse
|
9
|
Żuradzki T, Bystranowski P, Dranseika V. Discussions on Human Enhancement Meet Science: A Quantitative Analysis. SCIENCE AND ENGINEERING ETHICS 2025; 31:6. [PMID: 39907843 PMCID: PMC11799069 DOI: 10.1007/s11948-025-00531-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 01/21/2025] [Indexed: 02/06/2025]
Abstract
The analysis of citation flow from a collection of scholarly articles might provide valuable insights into their thematic focus and the genealogy of their main concepts. In this study, we employ a topic model to delineate a subcorpus of 1,360 papers representative of bioethical discussions on enhancing human life. We subsequently conduct an analysis of almost 11,000 references cited in that subcorpus to examine quantitatively, from a bird's-eye view, the degree of openness of this part of scholarship to the specialized knowledge produced in biosciences. Although almost half of the analyzed references point to journals classified as Natural Science and Engineering (NSE), we do not find strong evidence of the intellectual influence of recent discoveries in biosciences on discussions on human enhancement. We conclude that a large part of the discourse surrounding human enhancement is inflected with "science-fictional habits of mind." Our findings point to the need for a more science-informed approach in discussions on enhancing human life.
Collapse
Affiliation(s)
- Tomasz Żuradzki
- Jagiellonian University, Institute of Philosophy & Interdisciplinary Centre for Ethics, ul. Grodzka 52, Kraków, 31-044, Poland.
| | - Piotr Bystranowski
- Jagiellonian University, Interdisciplinary Centre for Ethics, ul. Grodzka 52, Kraków, 31-044, Poland
- Max Planck Institute for Research on Collective Goods, Bonn, Germany
| | - Vilius Dranseika
- Jagiellonian University, Interdisciplinary Centre for Ethics, ul. Grodzka 52, Kraków, 31-044, Poland
| |
Collapse
|
10
|
Hufana-Duran D, Chaikhun-Marcou T, Duran PG, Atabay EP, Nguyen HT, Atabay EC, Nguyen UT, Nguyen HT, Hiew MWH, Punyawai K, Ginting N, Parnpai R. Future of reproductive biotechnologies in water buffalo in Southeast Asian countries. Theriogenology 2025; 233:123-130. [PMID: 39613496 DOI: 10.1016/j.theriogenology.2024.11.016] [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: 08/05/2024] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 12/01/2024]
Abstract
The future of reproductive biotechnologies in water buffalo in Southeast Asian countries holds significant promise for enhancing genetic quality and productivity. Fixed-time artificial insemination remains the commonly used technology, with advances in assisted reproductive technologies (ART) such as in vitro embryo production (IVEP), embryo transfer (ET), and the use of sex-sorted sperm increasingly adopted to improve breeding efficiency. These technologies overcome traditional breeding limitations, such as low reproductive rates, genetic diversity constraints, and the production of sex-predetermined offspring. The application of multiple ovulation and embryo transfer (MOET) is constrained by poor embryo recovery in this livestock species. Somatic cell nuclear transfer (SCNT) offers great potential for producing sex-predetermined and genetically superior buffalo but requires further research to increase efficiency. Cryopreservation of buffalo genetics is bolstered by the establishment of Gene Banks. Challenges such as high costs, the need for skilled personnel, and infrastructure development remain constraints. Integration of genomic selection, automation, and expansion of ET programs are clear directions. Strengthening research and collaboration among Southeast Asian countries is essential to fully realize the benefits of these biotechnologies and ensure sustainable and profitable buffalo farming.
Collapse
Affiliation(s)
- Danilda Hufana-Duran
- Department of Agriculture-Philippine Carabao Center, Science City of Munoz, Nueva Ecija, 3120, Philippines
| | - Thuchadaporn Chaikhun-Marcou
- Obstetrics Gynecology Andrology and Animal Biotechnology Clinic, Faculty of Veterinary Medicine, Mahanakorn University of Technology, Bangkok, 10530, Thailand
| | - Peregrino G Duran
- Department of Agriculture-Philippine Carabao Center, Science City of Munoz, Nueva Ecija, 3120, Philippines
| | - Eufrocina P Atabay
- Department of Agriculture-Philippine Carabao Center, Science City of Munoz, Nueva Ecija, 3120, Philippines
| | - Hong Thi Nguyen
- Laboratory of Animal Biotechnology, Institute of Biotechnology, Vietnam Academy of Science and Technology, Hanoi, 100000, Viet Nam
| | - Edwin C Atabay
- Department of Agriculture-Philippine Carabao Center, Science City of Munoz, Nueva Ecija, 3120, Philippines
| | - Uoc Thi Nguyen
- Laboratory of Animal Biotechnology, Institute of Biotechnology, Vietnam Academy of Science and Technology, Hanoi, 100000, Viet Nam
| | - Hiep Thi Nguyen
- Laboratory of Animal Biotechnology, Institute of Biotechnology, Vietnam Academy of Science and Technology, Hanoi, 100000, Viet Nam
| | - Mark W H Hiew
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Kanchana Punyawai
- Agency for Science, Technology, and Research (A∗STAR), Institute of Molecular and Cell Biology (IMCB), Proteos, 138673, Singapore
| | - Nurzainah Ginting
- Animal Science Study Program, Universitas Sumatera Utara, Padang Bulan, Medan, 20155, Indonesia
| | - Rangsun Parnpai
- Embryo Technology and Stem Cell Research Center, School of Biotechnology, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima, 30000, Thailand.
| |
Collapse
|
11
|
Yoneyama Y, Zhang RR, Maezawa M, Masaki H, Kimura M, Cai Y, Adam M, Parameswaran S, Mizuno N, Bhadury J, Maezawa S, Ochiai H, Nakauchi H, Potter SS, Weirauch MT, Takebe T. Intercellular mRNA transfer alters the human pluripotent stem cell state. Proc Natl Acad Sci U S A 2025; 122:e2413351122. [PMID: 39841146 PMCID: PMC11789055 DOI: 10.1073/pnas.2413351122] [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: 07/10/2024] [Accepted: 12/07/2024] [Indexed: 01/23/2025] Open
Abstract
Intercellular transmission of messenger RNA (mRNA) is being explored in mammalian species using immortal cell lines. Here, we uncover an intercellular mRNA transfer phenomenon that allows for the adaptation and reprogramming of human primed pluripotent stem cells (hPSCs). This process is induced by the direct cell contact-mediated coculture with mouse embryonic stem cells under the condition impermissible for primed hPSC culture. Mouse-derived mRNA contents are transmitted into adapted hPSCs only in the coculture. Transfer-specific mRNA analysis shows the enrichment for divergent biological pathways involving transcription/translational machinery and stress-coping mechanisms, wherein such transfer is diminished when direct cell contacts are lost. After 5 d of coculture with mouse embryonic stem cells, surface marker analysis and global gene profiling confirmed that mRNA transfer-prone hPSC efficiently gains a naïve-like state. Furthermore, transfer-specific knockdown experiments targeting mouse-specific transcription factor-coding mRNAs in hPSC show that mouse-derived Tfcp2l1, Tfap2c, and Klf4 are indispensable for human naïve-like conversion. Thus, interspecies mRNA transfer triggers cellular reprogramming in mammalian cells. Our results support that episodic mRNA transfer can occur in cell cooperative and competitive processes, which provides a fresh perspective on understanding the roles of mRNA mobility for intra- and interspecies cellular communications.
Collapse
Affiliation(s)
- Yosuke Yoneyama
- Human Biology Research Unit, Institute of Integrated Research, Institute of Science Tokyo, Bunkyo-ku, Tokyo113-8510, Japan
- Department of Genome Biology, Graduate School of Medicine, Osaka University, Suita, Osaka565-0871, Japan
| | - Ran-Ran Zhang
- Division of Gastroenterology, Hepatology & Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
| | - Mari Maezawa
- Human Biology Research Unit, Institute of Integrated Research, Institute of Science Tokyo, Bunkyo-ku, Tokyo113-8510, Japan
| | - Hideki Masaki
- Stem Cell Therapy Division, Institute of Integrated Research, Institute of Science Tokyo, Bunkyo-ku, Tokyo113-8510, Japan
| | - Masaki Kimura
- Division of Gastroenterology, Hepatology & Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
| | - Yuqi Cai
- Division of Gastroenterology, Hepatology & Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
| | - Mike Adam
- Division of Gastroenterology, Hepatology & Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
| | - Sreeja Parameswaran
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
| | - Naoaki Mizuno
- Stem Cell Therapy Division, Institute of Integrated Research, Institute of Science Tokyo, Bunkyo-ku, Tokyo113-8510, Japan
| | - Joydeep Bhadury
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA94305
| | - So Maezawa
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Chiba278-8510, Japan
| | - Hiroshi Ochiai
- Division of Gene Expression Dynamics, Medical Institute of Bioregulation, Kyushu University, Fukuoka812-0054, Japan
| | - Hiromitsu Nakauchi
- Stem Cell Therapy Division, Institute of Integrated Research, Institute of Science Tokyo, Bunkyo-ku, Tokyo113-8510, Japan
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA94305
| | - S. Steven Potter
- Division of Gastroenterology, Hepatology & Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
| | - Matthew T. Weirauch
- Division of Gastroenterology, Hepatology & Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH45229-3039
| | - Takanori Takebe
- Human Biology Research Unit, Institute of Integrated Research, Institute of Science Tokyo, Bunkyo-ku, Tokyo113-8510, Japan
- Department of Genome Biology, Graduate School of Medicine, Osaka University, Suita, Osaka565-0871, Japan
- Division of Gastroenterology, Hepatology & Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH45229-3039
- Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children’s Hospital Medical Center, Cincinnati, OH45229-3039
- World Premier International Research Center Initiative Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Suita, Osaka565-0871, Japan
| |
Collapse
|
12
|
Gallardo MD, Girard M, Portiansky EL, Goya RG. Oct4, Sox2, Klf4, c-My (OSKM) gene therapy in the hypothalamus prolongs fertility and ovulation in female rats. Aging (Albany NY) 2025; 17:161-169. [PMID: 39864409 PMCID: PMC11810065 DOI: 10.18632/aging.206191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 01/15/2025] [Indexed: 01/28/2025]
Abstract
In middle-aged (MA) female rats, we have demonstrated that intrahypothalamic gene therapy for insulin-like growth factor-I (IGF-I) extends the regular cyclicity of the animals beyond 10 months (the age at which MA rats stop ovulating). Here, we implemented long-term OSKM gene therapy in the hypothalamus of young female rats. The main goal was to extend fertility in the treated animals. We constructed an adenovector that harbors the GFP gene as well as 4 Yamanaka genes. An adenovector that only carries the gene for GFP or DsRed was used as control. At 4 months of age 12 female rats received an intrahypothalamic injection of our OSKM vector (treated rats); 12 control rats received a vector expressing a marker gene (control rats). At 9.3 months of age control and treated rats were mated with young males. A group of 12 young intact female rats was also mated. The rate of pregnancy recorded was 83%, 8.3 and 25% for young, MA control and MA treated animals, respectively. Pup body weight (BW) at weaning was significantly higher in the MA OSKM rats than in MA controls. At the age of estropause (10 months), OSKM treated females still showed regular estrous cycles. The particular significance of the present results is that, for the first time, it is shown that long-term OSKM gene therapy in the hypothalamus is able to extend the functionality of such a complex system as the hypothalamo-pituitary-ovarian axis.
Collapse
Affiliation(s)
- Maria D. Gallardo
- School of Medicine, National University of La Plata (UNLP), La Plata, Argentina
| | - Mauricio Girard
- School of Medicine, National University of La Plata (UNLP), La Plata, Argentina
| | - Enrique L. Portiansky
- Image Analysis lab; School of Veterinary Sciences, National University of La Plata, La Plata, Argentina
| | - Rodolfo G. Goya
- School of Medicine, National University of La Plata (UNLP), La Plata, Argentina
- Biomedical Research and Longevity Society, Fort Lauderdale, FL 33309, USA
| |
Collapse
|
13
|
Nesiyama TNG, Sangalli JR, De Bem THC, Recchia K, Martins SMMK, de Andrade AFC, Ferst JG, Almeida GHDR, Marques MG, Dória RGS, Carregaro AB, Feliciano MAR, Miglino MA, Bressan FF, Perecin F, da Silveira JC, Smith LC, Bordignon V, Meirelles FV. Swine clones: potential application for animal production and animal models. Anim Reprod 2025; 22:e20240037. [PMID: 39867300 PMCID: PMC11758785 DOI: 10.1590/1984-3143-ar2024-0037] [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: 04/15/2024] [Accepted: 10/10/2024] [Indexed: 01/28/2025] Open
Abstract
Somatic cell nuclear transfer (SCNT), or cloning, is used to reprogram cells and generate genetically identical embryos and animals. However, the cloning process is inefficient, limiting its application to producing valuable animals. In swine, cloning is mainly utilized to produce genetically modified animals. Indeed, recombinant DNA technologies have evolved considerably in recent years, with homologous recombination and gene editing technologies becoming more efficient and capable of recombining both alleles in a single cell. The selection of appropriate cells and their use as nuclear donors for SCNT is the most common method for generating edited and genetically modified animals for commercial and research purposes. This article reviews current applications of swine cloning and shares our personal experiences with the procedure in this species.
Collapse
Affiliation(s)
| | - Juliano Rodrigues Sangalli
- Faculdade de Zootecnia e Engenharia de Alimentos – FZEA, Universidade de São Paulo – USP, Pirassununga, SP, Brasil
| | - Tiago Henrique Camara De Bem
- Faculdade de Zootecnia e Engenharia de Alimentos – FZEA, Universidade de São Paulo – USP, Pirassununga, SP, Brasil
| | - Kaiana Recchia
- Faculdade de Medicina Veterinária e Zootecnia – FMVZ, Universidade de São Paulo – USP, Pirassununga, SP, Brasil
| | | | | | - Juliana Germano Ferst
- Faculdade de Zootecnia e Engenharia de Alimentos – FZEA, Universidade de São Paulo – USP, Pirassununga, SP, Brasil
| | | | | | - Renata Gebara Sampaio Dória
- Faculdade de Zootecnia e Engenharia de Alimentos – FZEA, Universidade de São Paulo – USP, Pirassununga, SP, Brasil
| | - Adriano Bonfim Carregaro
- Faculdade de Zootecnia e Engenharia de Alimentos – FZEA, Universidade de São Paulo – USP, Pirassununga, SP, Brasil
| | | | - Maria Angélica Miglino
- Faculdade de Medicina Veterinária e Zootecnia – FMVZ, Universidade de São Paulo – USP, Pirassununga, SP, Brasil
| | - Fabiana Fernandes Bressan
- Faculdade de Zootecnia e Engenharia de Alimentos – FZEA, Universidade de São Paulo – USP, Pirassununga, SP, Brasil
| | - Felipe Perecin
- Faculdade de Zootecnia e Engenharia de Alimentos – FZEA, Universidade de São Paulo – USP, Pirassununga, SP, Brasil
| | - Juliano Coelho da Silveira
- Faculdade de Zootecnia e Engenharia de Alimentos – FZEA, Universidade de São Paulo – USP, Pirassununga, SP, Brasil
| | - Lawrence Charles Smith
- Faculté de Médecine Vétérinaire – FMV, Université de Montréal – UdeM, Montréal, Quebec, Canada
| | - Vilceu Bordignon
- McGill Faculty of Agricultural and Environmental Science – FAES, McGill University, Montréal, Quebec, Canada
| | - Flávio Vieira Meirelles
- Faculdade de Zootecnia e Engenharia de Alimentos – FZEA, Universidade de São Paulo – USP, Pirassununga, SP, Brasil
| |
Collapse
|
14
|
Liu HJ, Oh SJWY, Tay NL, Lim CY, Hsu CD, Chua DHH, Teo WKL, Loh YH, Ng SC. Morphokinetic Analyses of Fishing Cat-Domestic Cat Interspecies Somatic Cell Nuclear Transfer Embryos Through A Time-Lapse System. Animals (Basel) 2025; 15:148. [PMID: 39858147 PMCID: PMC11758314 DOI: 10.3390/ani15020148] [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: 12/06/2024] [Revised: 12/30/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
A time-lapse live embryo monitoring system provides a powerful approach to recording dynamic developmental events of cultured embryos in detail. By obtaining continuous short-interval images, blastocyst formation can be predicted and embryos can be selected. The objective of this study was to investigate the morphokinetic parameters of fishing cat-domestic cat interspecies somatic cell nuclear transfer (iSCNT) embryos from one-cell to blastocyst stages, and in particular, the cleavage patterns of the first division in iSCNT and IVF embryos, as these play a central role in euploidy. Domestic cat in vitro fertilization (IVF) embryos were set up as controls. The results show that morula and blastocyst development rates were significantly lower in the iSCNT embryos compared to their IVF counterparts. All earlier time points of embryonic development before the onset of blastulation in the iSCNT embryos were significantly delayed when compared with their IVF counterparts. In iSCNT, normal embryos (defined as those that developed to the blastocyst stage) took a longer time to reach the morula stage, and these morulas were more likely to undergo compaction, compared to their arrested embryo counterparts. Direct cleavage in the first division is a morphological aberration, and was seen with greater prevalence in iSCNT embryos than control IVF embryos; these aberrant embryos displayed a significantly lower blastocyst development rate than embryos that had undergone normal cleavage. In conclusion, the morphokinetic parameters of fishing cat-domestic cat iSCNT embryos at early stages could be used to predict their potential for development to the blastocyst stage. A time-lapse imaging system is potentially a powerful tool for selecting early embryos with developmental potential for transfer, and hence, for improving feline iSCNT efficiency.
Collapse
Affiliation(s)
- Hai-Jun Liu
- Endangered Species Conservation via Assisted Reproduction (ESCAR) Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore; (H.-J.L.); (S.J.W.Y.O.); (N.L.T.); (C.Y.L.); (W.K.L.T.); (Y.-H.L.)
| | - Serena Jocelyn Wai Yin Oh
- Endangered Species Conservation via Assisted Reproduction (ESCAR) Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore; (H.-J.L.); (S.J.W.Y.O.); (N.L.T.); (C.Y.L.); (W.K.L.T.); (Y.-H.L.)
| | - Nicole Liling Tay
- Endangered Species Conservation via Assisted Reproduction (ESCAR) Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore; (H.-J.L.); (S.J.W.Y.O.); (N.L.T.); (C.Y.L.); (W.K.L.T.); (Y.-H.L.)
| | - Christina Yingyan Lim
- Endangered Species Conservation via Assisted Reproduction (ESCAR) Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore; (H.-J.L.); (S.J.W.Y.O.); (N.L.T.); (C.Y.L.); (W.K.L.T.); (Y.-H.L.)
| | - Chia-Da Hsu
- Mandai Wildlife Group, 80 Mandai Lake Road, Singapore 729826, Singapore; (C.-D.H.); (D.H.H.C.)
| | - Delia Hwee Hoon Chua
- Mandai Wildlife Group, 80 Mandai Lake Road, Singapore 729826, Singapore; (C.-D.H.); (D.H.H.C.)
| | - Winnie Koon Lay Teo
- Endangered Species Conservation via Assisted Reproduction (ESCAR) Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore; (H.-J.L.); (S.J.W.Y.O.); (N.L.T.); (C.Y.L.); (W.K.L.T.); (Y.-H.L.)
| | - Yuin-Han Loh
- Endangered Species Conservation via Assisted Reproduction (ESCAR) Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore; (H.-J.L.); (S.J.W.Y.O.); (N.L.T.); (C.Y.L.); (W.K.L.T.); (Y.-H.L.)
- Cell Fate Engineering and Therapeutics Laboratory, Cell Biology and Therapies Division, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, 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
- Endangered Species Conservation via Assisted Reproduction (ESCAR) Lab, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore; (H.-J.L.); (S.J.W.Y.O.); (N.L.T.); (C.Y.L.); (W.K.L.T.); (Y.-H.L.)
- 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
| |
Collapse
|
15
|
Jiang S, Li H, Zhang L, Mu W, Zhang Y, Chen T, Wu J, Tang H, Zheng S, Liu Y, Wu Y, Luo X, Xie Y, Ren J. Generic Diagramming Platform (GDP): a comprehensive database of high-quality biomedical graphics. Nucleic Acids Res 2025; 53:D1670-D1676. [PMID: 39470721 PMCID: PMC11701665 DOI: 10.1093/nar/gkae973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/30/2024] [Accepted: 10/11/2024] [Indexed: 10/30/2024] Open
Abstract
High-quality schematic illustrations are fundamental to the publication of scientific achievements in biomedical research, which are crucial for effectively conveying complex biomedical concepts. However, creating such illustrations remains challenging for many researchers due to the need to devote a significant amount of time and effort to accomplish it. To address this need, we present the Generic Diagramming Platform (GDP, https://BioGDP.com), a comprehensive database of professionally crafted biomedical graphics (bio-graphics). Currently, GDP houses 7 562 high-quality bio-graphics, meticulously categorized into 10 major and 77 minor categories. To increase the design efficiency, GDP provides 204 customizable templates derived from an extensive review of over 2000 literature and 7 textbooks. With the interactive drawing platform and user-friendly web interface implemented in GDP, these resources can facilitate the efficient generation of publication-ready illustrations for the biomedical community. Additionally, GDP incorporates a collaborative submission system, allowing researchers to contribute their artwork, fostering a growing diagramming ecosystem, and ensuring continuous database expansion. Overall, we believe that GDP will serve as an invaluable platform, significantly enhancing the efficiency and quality of scientific illustration for biomedical researchers.
Collapse
Affiliation(s)
- Shuai Jiang
- School of Life Sciences, State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Huiqin Li
- School of Life Sciences, State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Luowanyue Zhang
- School of Life Sciences, State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Weiping Mu
- School of Life Sciences, State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Ya Zhang
- School of Life Sciences, State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Tianjian Chen
- School of Life Sciences, State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Jingxing Wu
- School of Life Sciences, State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Haoyun Tang
- School of Life Sciences, State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Shuxin Zheng
- School of Life Sciences, State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Yifei Liu
- School of Life Sciences, State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Yaxuan Wu
- School of Life Sciences, State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| | - Xiaotong Luo
- Guangdong Institute of Gastroenterology, Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510060, China
| | - Yubin Xie
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Jian Ren
- School of Life Sciences, State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University, Guangzhou 510060, China
| |
Collapse
|
16
|
Kamada Y, Yamaji K, Ushigome N, Ito D, Wakayama S, Hiraoka K, Hayashi M, Kawai K, Wakayama T. Method for long-term room temperature storage of mouse freeze-dried sperm. Sci Rep 2025; 15:303. [PMID: 39747170 PMCID: PMC11695616 DOI: 10.1038/s41598-024-83350-2] [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: 07/24/2024] [Accepted: 12/13/2024] [Indexed: 01/04/2025] Open
Abstract
Permanent preservation of genetic resources may be indispensable for the future of humanity. This requires liquid nitrogen, as is the case for preserving animal sperm. However, this technique is expensive and poses a risk of irrecoverable sample loss on non-replenishment of liquid nitrogen in case of natural disasters. In this study, we demonstrate that lyophilization may be used as a reliable method for long-term preservation of mouse sperm at room temperature. Sperm from four mouse strains were freeze-dried and stored in a non-temperature controlled room for 5-6 years. Although the ability of the stored sperm to activate oocytes had diminished slightly, healthy offspring were obtained by artificially activating the oocytes after sperm injection. Moreover, the birth rate did not decrease even after ≤ 6 years of storage. Furthermore, owing to its low cost, safety, and ease of storage at any location, we believe that this method could be a major mode of preserving mammalian genetic resources in the future.
Collapse
Affiliation(s)
- Yuko Kamada
- Faculty of Life and Environmental Science, University of Yamanashi, Yamanashi, Japan
- Department of Reproductive Medicine, Kameda Medical Center (Kameda General Hospital), Chiba, Japan
| | - Kango Yamaji
- Faculty of Life and Environmental Science, University of Yamanashi, Yamanashi, Japan
| | - Natsuki Ushigome
- Faculty of Life and Environmental Science, University of Yamanashi, Yamanashi, Japan
| | - Daiyu Ito
- Faculty of Life and Environmental Science, University of Yamanashi, Yamanashi, Japan
| | - Sayaka Wakayama
- Advanced Biotechnology Center, University of Yamanashi, Yamanashi, Japan
| | - Kenichiro Hiraoka
- Department of Reproductive Medicine, Kameda Medical Center (Kameda General Hospital), Chiba, Japan
- Department of Reproductive Medicine, Kameda IVF Clinic Makuhari, Chiba, Japan
| | - Masaru Hayashi
- Department of Reproductive Medicine, Kameda Medical Center (Kameda General Hospital), Chiba, Japan
| | - Kiyotaka Kawai
- Department of Reproductive Medicine, Kameda Medical Center (Kameda General Hospital), Chiba, Japan
- Department of Reproductive Medicine, Kameda IVF Clinic Makuhari, Chiba, Japan
| | - Teruhiko Wakayama
- Faculty of Life and Environmental Science, University of Yamanashi, Yamanashi, Japan.
- Advanced Biotechnology Center, University of Yamanashi, Yamanashi, Japan.
| |
Collapse
|
17
|
Aizawa E, Peters AHFM, Wutz A. In vitro gametogenesis: Towards competent oocytes: Limitations and future improvements for generating oocytes from pluripotent stem cells in culture. Bioessays 2025; 47:e2400106. [PMID: 39498732 DOI: 10.1002/bies.202400106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 09/25/2024] [Accepted: 10/10/2024] [Indexed: 11/07/2024]
Abstract
Production of oocytes from pluripotent cell cultures in a dish represents a new paradigm in stem cell and developmental biology and has implications for how we think about life. The spark of life for the next generation occurs at fertilization when sperm and oocyte fuse. In animals, gametes are the only cells that transmit their genomes to the next generation. Oocytes contain in addition a large cytoplasm with factors that direct embryonic development. Reconstitution of mouse oocyte and embryonic development in culture provides experimental opportunities and facilitates an unprecedented understanding of molecular mechanisms. However, the application of in vitro gametogenesis to reproductive medicine or infertility treatment remains challenging. One significant concern is the quality of in vitro-derived oocytes. Here, we review the current understanding and identify limitations in generating oocytes in vitro. From this basis, we explore opportunities for future improvements of the in vitro approach to generating high-quality oocytes.
Collapse
Affiliation(s)
- Eishi Aizawa
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology, ETH Zurich, Zurich, Switzerland
- RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Antoine H F M Peters
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
- Faculty of Sciences, University of Basel, Basel, Switzerland
| | - Anton Wutz
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
18
|
Conboy I, Sviercovich A. Only Some Paths Lead to Longer Healthier Life and How to Find Them. Rejuvenation Res 2024; 27:v-vii. [PMID: 39657033 DOI: 10.1089/rej.2024.111424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024] Open
|
19
|
Müller GA, Müller TD. A "poly-matter network" conception of biological inheritance. Genetica 2024; 152:211-230. [PMID: 39425866 PMCID: PMC11541361 DOI: 10.1007/s10709-024-00216-1] [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: 02/23/2024] [Accepted: 09/26/2024] [Indexed: 10/21/2024]
Abstract
Here we intend to shift the "DNA- and information-centric" conception of biological inheritance, with the accompanying exclusion of any non-DNA matter, to a "poly-matter network" framework which, in addition to DNA, considers the action of other cellular membranous constituents. These cellular structures, in particular organelles and plasma membranes, express "landscapes" of specific topologies at their surfaces, which may become altered in response to certain environmental factors. These so-called "membranous environmental landscapes" (MELs), which replicate by self-organization / autopoiesis rather than self-assembly, are transferred from donor to acceptor cells by various - vesicular and non-vesicular - mechanisms and exert novel features in the acceptor cells. The "DNA-centric" conception may be certainly explanatorily sufficient for the transfer of heritable phenotype variation to acceptor cells following the copying of DNA in donor cells and thereby for the phenomenon of biological inheritance of traits. However, it is not causally sufficient. With the observation of phenotype variation, as initially manifested during bacterial transformation, the impact of environmental factors, such as nutrition and stress, in the differential regulation of gene expression has been widely accepted and resulted in intense efforts to resolve the underlying epigenetic mechanisms. However, these are explained under a conceptual frame where the DNA (and associated proteins) are the only matter of inheritance. In contrast, it is our argumentation that inheritance can only be adequately understood as the transfer of DNA in concert with non-DNA matter in a "poly-matter network" conception. The adequate inclusion of the transfer of non-DNA matter is still a desideratum of future genetic research, which may pave the way for the experimental elucidation not only of how DNA and membrane matter act in concert to enable the inheritance of innate traits, but also whether they interact for that of acquired biological traits. Moreover, the "poly-matter network" conception may open new perspectives for an understanding of the pathogenesis of "common complex" diseases.
Collapse
Affiliation(s)
- Günter A Müller
- Institute of Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC) at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764, Oberschleissheim, Germany.
- Biology and Technology Studies Institute Munich (BITSIM), Lappenweg 16, 80939, Munich, Germany.
- Media, Culture and Society, Department of Media Studies, Faculty of Arts and Humanities, University Paderborn, Warburger Str. 100, 33098, Paderborn, Germany.
| | - Timo D Müller
- Institute of Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC) at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764, Oberschleissheim, Germany
- Walther-Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
20
|
Jiang Y, Harberts J, Assadi A, Chen Y, Spatz JP, Duan W, Nisbet DR, Voelcker NH, Elnathan R. The Roles of Micro- and Nanoscale Materials in Cell-Engineering Systems. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2410908. [PMID: 39401098 DOI: 10.1002/adma.202410908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/13/2024] [Indexed: 11/29/2024]
Abstract
Customizable manufacturing of ex vivo cell engineering is driven by the need for innovations in the biomedical field and holds substantial potential for addressing current therapeutic challenges; but it is still only in its infancy. Micro- and nanoscale-engineered materials are increasingly used to control core cell-level functions in cellular engineering. By reprogramming or redirecting targeted cells for extremely precise functions, these advanced materials offer new possibilities. This influences the modularity of cell reprogramming and reengineering, making these materials part of versatile and emerging technologies. Here, the roles of micro- and nanoscale materials in cell engineering are highlighted, demonstrating how they can be adaptively controlled to regulate cellular reprogramming and core cell-level functions, including differentiation, proliferation, adhesion, user-defined gene expression, and epigenetic changes. The current reprogramming routes used to achieve pluripotency from somatic cells and the significant potential of induced pluripotent stem cell technology for translational biomedical research are covered. Recent advances in nonviral intracellular delivery modalities for cell reprogramming and their constraints are evaluated. This paper focuses on emerging physical and combinatorial approaches of intracellular delivery for cell engineering, revealing the capabilities and limitations of these routes. It is showcased how these programmable materials are continually being explored as customizable tools for inducing biophysical stimulation. Harnessing the power of micro- and nanoscale-engineered materials will be a step change in the design of cell engineering, producing a suite of powerful tools for addressing potential future challenges in therapeutic cell engineering.
Collapse
Affiliation(s)
- Yuan Jiang
- Faculty of Health, School of Medicine, Deakin University, Waurn Ponds, Victoria, 3216, Australia
- The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Waurn Ponds, Victoria, 3216, Australia
| | - Jann Harberts
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Victoria, Clayton, 3168, Australia
| | - Artin Assadi
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Victoria, Clayton, 3168, Australia
| | - Yaping Chen
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Zhejiang, 325000, China
| | - Joachim P Spatz
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, 69120, Heidelberg, Germany
- Institute for Molecular Systems Engineering (IMSE), Heidelberg University, 69120, Heidelberg, Germany
- Max Planck School Matter to Life, Max Planck Schools, 69120, Heidelberg, Germany
| | - Wei Duan
- Faculty of Health, School of Medicine, Deakin University, Waurn Ponds, Victoria, 3216, Australia
| | - David R Nisbet
- The Graeme Clark Institute, University of Melbourne, Parkville, Victoria, 3010, Australia
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Melbourne, Parkville, Victoria, 3010, Australia
- Medical School, Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Melbourne, Parkville, VIC, 3010, Australia
| | - Nicolas H Voelcker
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Victoria, Clayton, 3168, Australia
| | - Roey Elnathan
- Faculty of Health, School of Medicine, Deakin University, Waurn Ponds, Victoria, 3216, Australia
- The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Waurn Ponds, Victoria, 3216, Australia
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Victoria, Clayton, 3168, Australia
- Institute for Frontier Materials, Deakin University, Waurn Ponds, Victoria, 3216, Australia
| |
Collapse
|
21
|
Punetha M, Saini S, Choudhary S, Sharma S, Bala R, Kumar P, Sharma RK, Yadav PS, Datta TK, Kumar D. Establishment of CRISPR-Cas9 ribonucleoprotein mediated MSTN gene edited pregnancy in buffalo: Compare cells transfection and zygotes electroporation. Theriogenology 2024; 229:158-168. [PMID: 39178617 DOI: 10.1016/j.theriogenology.2024.08.027] [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: 06/18/2024] [Revised: 08/20/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024]
Abstract
Genome editing is recognized as a powerful tool in agriculture and research, enhancing our understanding of genetic function, diseases, and productivity. However, its progress in buffaloes has lagged behind other mammals due to several challenges, including long gestational periods, single pregnancies, and high raising costs. In this study, we aimed to generate MSTN-edited buffaloes, known for their distinctive double-muscling phenotype, as a proof of concept. To meet our goal, we used somatic cell nuclear transfer (SCNT) and zygotic electroporation (CRISPR-EP) technique. For this, we firstly identified the best transfection method for introduction of RNP complex into fibroblast which was further used for SCNT. For this, we compared the transfection, cleavage efficiency and cell viability of nucleofection and lipofection in adult fibroblasts. The cleavage, transfection efficiency and cell viability of nucleofection group was found to be significantly (P ≤ 0.05) higher than lipofection group. Four MSTN edited colony were generated using nucleofection, out of which three colonies was found to be biallelic and one was monoallelic. Further, we compared the efficacy, embryonic developmental potential and subsequent pregnancy outcome of SCNT and zygotic electroporation. The blastocyst rate of electroporated group was found to be significantly (P ≤ 0.05) higher than SCNT group. However, the zygotic electroporation group resulted into two pregnancies which were confirmed to be MSTN edited. Since, the zygotic electroporation does not require complex micromanipulation techniques associated with SCNT, it has potential for facilitating the genetic modification in large livestock such as buffaloes. The present study lays the basis for inducing genetic alternation with practical or biological significance.
Collapse
Affiliation(s)
- Meeti Punetha
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar-125001, Haryana, India
| | - Sheetal Saini
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar-125001, Haryana, India
| | - Suman Choudhary
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar-125001, Haryana, India
| | - Surabhi Sharma
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar-125001, Haryana, India
| | - Renu Bala
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar-125001, Haryana, India
| | - Pradeep Kumar
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar-125001, Haryana, India
| | - R K Sharma
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar-125001, Haryana, India
| | - P S Yadav
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar-125001, Haryana, India
| | - T K Datta
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar-125001, Haryana, India
| | - Dharmendra Kumar
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar-125001, Haryana, India.
| |
Collapse
|
22
|
Parmar B, Bhatia D. Small Molecular Approaches for Cellular Reprogramming and Tissue Engineering: Functions as Mediators of the Cell Signaling Pathway. Biochemistry 2024; 63:2542-2556. [PMID: 39312802 DOI: 10.1021/acs.biochem.4c00427] [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: 09/25/2024]
Abstract
Utilizing induced pluripotent stem cells (iPSCs) in drug screening and cell replacement therapy has emerged as a method with revolutionary applications. With the advent of patient-specific iPSCs and the subsequent development of cells that exhibit disease phenotypes, the focus of medication research will now shift toward the pathology of human diseases. Regular iPSCs can also be utilized to generate cells that assess the negative impacts of medications. These cells provide a much more precise and cost-efficient approach compared to many animal models. In this review, we explore the utilization of small-molecule drugs to enhance the growth of iPSCs and gain insights into the process of reprogramming. We mainly focus on the functions of small molecules in modulating different signaling pathways, thereby modulating cell fate. Understanding the way small molecule drugs interact with iPSC technology has the potential to significantly enhance the understanding of physiological pathways in stem cells and practical applications of iPSC-based therapy and screening systems, revolutionizing the treatment of diseases.
Collapse
Affiliation(s)
- Bhagyesh Parmar
- Department of Biological Sciences and Engineering, Indian Institute of Technology, Palaj, Gandhinagar 382355, India
| | - Dhiraj Bhatia
- Department of Biological Sciences and Engineering, Indian Institute of Technology, Palaj, Gandhinagar 382355, India
| |
Collapse
|
23
|
Montoliu L. Ethical aspects associated with genome alteration techniques applied to animal reproduction research. Reprod Domest Anim 2024; 59 Suppl 3:e14670. [PMID: 39396877 DOI: 10.1111/rda.14670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 10/15/2024]
Abstract
The revolution in biology triggered by the different genome-editing tools has of course arrived to the research field of animal reproduction. Yeast meganucleases, zinc-finger nucleases, TALEN and, particularly, the several generations of CRISPR tools have landed in animal reproduction thereby providing novel strategies to optimize or modify some of the features and capabilities of the recipient animals. All these genome-editing proposals and activities are associated with ethical considerations regarding how those planned genome alterations might affect important animal welfare issues. The ethical dimension of all these genome editing must be seriously considered. Hence, all ethical aspects bound to any given genome-edited allele in animals should be discussed in order to ensure that we are maximizing benefits and reducing any potential risk or negative considerations of these modifications. In this review, I will summarize some of the experiments reported aiming to investigate or improve animal reproduction and I will address the ethics issues that should also be considered.
Collapse
Affiliation(s)
- Lluis Montoliu
- Department of Molecular and Cellular Biology, National Centre for Biotechnology (CNB-CSIC), Madrid, Spain
- Rare Diseases Networking Biomedical Research Centre (CIBERER-ISCIII), Madrid, Spain
| |
Collapse
|
24
|
Radwan A, Eccleston J, Sabag O, Marcus H, Sussman J, Ouro A, Rahamim M, Azagury M, Azria B, Stanger BZ, Cedar H, Buganim Y. Transdifferentiation occurs without resetting development-specific DNA methylation, a key determinant of full-function cell identity. Proc Natl Acad Sci U S A 2024; 121:e2411352121. [PMID: 39292740 PMCID: PMC11441492 DOI: 10.1073/pnas.2411352121] [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: 07/02/2024] [Accepted: 08/21/2024] [Indexed: 09/20/2024] Open
Abstract
A number of studies have demonstrated that it is possible to directly convert one cell type to another by factor-mediated transdifferentiation, but in the vast majority of cases, the resulting reprogrammed cells are unable to maintain their new cell identity for prolonged culture times and have a phenotype only partially similar to their endogenous counterparts. To better understand this phenomenon, we developed an analytical approach for better characterizing trans-differentiation-associated changes in DNA methylation, a major determinant of long-term cell identity. By examining various models of transdifferentiation both in vitro and in vivo, our studies indicate that despite convincing expression changes, transdifferentiated cells seem unable to alter their original developmentally mandated methylation patterns. We propose that this blockage is due to basic developmental limitations built into the regulatory sequences that govern epigenetic programming of cell identity. These results shed light on the molecular rules necessary to achieve complete somatic cell reprogramming.
Collapse
Affiliation(s)
- Ahmed Radwan
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hebrew University Medical School, Jerusalem91120, Israel
| | - Jason Eccleston
- Department of Medicine and Cell, The Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA19104
- Department of Development Biology, The Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA19104
| | - Ofra Sabag
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hebrew University Medical School, Jerusalem91120, Israel
| | - Howard Marcus
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hebrew University Medical School, Jerusalem91120, Israel
| | - Jonathan Sussman
- Department of Medicine and Cell, The Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA19104
- Department of Development Biology, The Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA19104
| | - Alberto Ouro
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hebrew University Medical School, Jerusalem91120, Israel
| | - Moran Rahamim
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hebrew University Medical School, Jerusalem91120, Israel
| | - Meir Azagury
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hebrew University Medical School, Jerusalem91120, Israel
| | - Batia Azria
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hebrew University Medical School, Jerusalem91120, Israel
| | - Ben Z. Stanger
- Department of Medicine and Cell, The Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA19104
- Department of Development Biology, The Institute for Regenerative Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA19104
| | - Howard Cedar
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hebrew University Medical School, Jerusalem91120, Israel
| | - Yosef Buganim
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Hebrew University Medical School, Jerusalem91120, Israel
| |
Collapse
|
25
|
Rodriguez-Villamil P, Beaton BP, Krisher RL. Gene editing in livestock: innovations and applications. Anim Reprod 2024; 21:e20240054. [PMID: 39372257 PMCID: PMC11452096 DOI: 10.1590/1984-3143-ar2024-0054] [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: 04/29/2024] [Accepted: 08/05/2024] [Indexed: 10/08/2024] Open
Abstract
Gene editing technologies have revolutionized the field of livestock breeding, offering unprecedented opportunities to enhance animal welfare, productivity, and sustainability. This paper provides a comprehensive review of recent innovations and applications of gene editing in livestock, exploring the diverse applications of gene editing in livestock breeding, as well as the regulatory and ethical considerations, and the current challenges and prospects of the technology in the industry. Overall, this review underscores the transformative potential of gene editing in livestock breeding and its pivotal role in shaping the future of agriculture and biomedicine.
Collapse
|
26
|
Ju BH, Kim YJ, Park YB, Kim BH, Kim MK. Evaluation of conical 9 well dish on bovine oocyte maturation and subsequent embryonic development. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2024; 66:936-948. [PMID: 39398310 PMCID: PMC11466740 DOI: 10.5187/jast.2024.e68] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/26/2024] [Accepted: 07/01/2024] [Indexed: 08/22/2024]
Abstract
The Conical 9 well dish (C9 well dish) is characterized by a decreasing cross-sectional area towards the base. This design was hypothesized to enhance embryonic development by emulating the in vivo physical environment through density modulation. Comparative analyses revealed no significant difference in nuclear maturation rates between the C9 well dish and the 5-well dish. Reactive oxygen species (ROS) generation was lower in the C9 well dish compared to the 5-well dish; however, this difference was not statistically significant. On the second day of in vitro culture, the cleavage rate in the C9 well dish was 4.66% higher, although not statistically significant, and the rates of blastocyst development were similar across both dishes. No significant differences were observed in the intracellular levels of glutathione (GSH) and ROS, as well as in the total cell number within the blastocysts between the dish types. The expression of mitogen-related factors, TGFα and IGF-1, in the blastocysts was consistent between the dishes. However, PDGFβ expression was significantly lower in the C9 well dish compared to the 35 mm petri dish. Similarly, the expression of the apoptosis factor Bax/Bcl2l2 showed no significant differences between the two dishes. Despite the marked difference in PDGFβ expression, its impact on blastocyst formation appeared negligible. The study also confirmed the feasibility of culturing a small number of oocytes per donor, collected via Ovum Pick-Up (OPU), with reduced volumes of culture medium and mineral oil, thus offering economic advantages. In conclusion, the present study indicates that the C9 well dish is effective for in vitro development of a small quantity of oocytes and embryos, presenting it as a viable alternative to traditional cell culture dishes.
Collapse
Affiliation(s)
- Byung Hyun Ju
- Division of Animal and Dairy Science,
College of Agriculture and Life Science, Chungnam National
University, Daejeon 34134, Korea
- MK biotech Inc., Daejeon
34134, Korea
| | - You Jin Kim
- Department of Obstetrics &
Gynecology, Chungnam National University Hospital, Daejeon
34134, Korea
| | - Youn Bae Park
- Division of Animal and Dairy Science,
College of Agriculture and Life Science, Chungnam National
University, Daejeon 34134, Korea
- MK biotech Inc., Daejeon
34134, Korea
| | - Byeong Ho Kim
- Division of Animal and Dairy Science,
College of Agriculture and Life Science, Chungnam National
University, Daejeon 34134, Korea
- MK biotech Inc., Daejeon
34134, Korea
| | - Min Kyu Kim
- Division of Animal and Dairy Science,
College of Agriculture and Life Science, Chungnam National
University, Daejeon 34134, Korea
- MK biotech Inc., Daejeon
34134, Korea
| |
Collapse
|
27
|
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
|
28
|
Wei Q, Xu Y, Cui G, Sun J, Su Z, Kou X, Zhao Y, Cao S, Li W, Xu Y, Gao S. Male-pronuclei-specific granulin facilitates somatic cells reprogramming via mitigating excessive cell proliferation and enhancing lysosomal function. J Cell Physiol 2024; 239:e31295. [PMID: 38747637 DOI: 10.1002/jcp.31295] [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: 11/01/2023] [Revised: 04/05/2024] [Accepted: 04/30/2024] [Indexed: 08/15/2024]
Abstract
Critical reprogramming factors resided predominantly in the oocyte or male pronucleus can enhance the efficiency or the quality of induced pluripotent stem cells (iPSCs) induction. However, few reprogramming factors exist in the male pronucleus had been verified. Here, we demonstrated that granulin (Grn), a factor enriched specifically in male pronucleus, can significantly improve the generation of iPSCs from mouse fibroblasts. Grn is highly expressed on Day 1, Day 3, Day 14 of reprogramming induced by four Yamanaka factors and functions at the initial stage of reprogramming. Transcriptome analysis indicates that Grn can promote the expression of lysosome-related genes, while inhibit the expression of genes involved in DNA replication and cell cycle at the early reprogramming stage. Further verification determined that Grn suppressed cell proliferation due to the arrest of cell cycle at G2/M phase. Moreover, ectopic Grn can enhance the lysosomes abundance and rescue the efficiency reduction of reprogramming resulted from lysosomal protease inhibition. Taken together, we conclude that Grn serves as an activator for somatic cell reprogramming through mitigating cell hyperproliferation and promoting the function of lysosomes.
Collapse
Affiliation(s)
- Qingqing Wei
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian, China
| | - Yanwen Xu
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian, China
| | - Guina Cui
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian, China
| | - Jiatong Sun
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian, China
| | - Zhongqu Su
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian, China
| | - Xiaochen Kou
- Clinical and Translation Research Center of Shanghai First Maternity & Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yanhong Zhao
- Clinical and Translation Research Center of Shanghai First Maternity & Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Suyuan Cao
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian, China
| | - Wenhui Li
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian, China
| | - Yiliang Xu
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Taian, China
| | - Shaorong Gao
- Clinical and Translation Research Center of Shanghai First Maternity & Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| |
Collapse
|
29
|
Moura MT. Genome-Scale Analyses Reveal Roadblocks to Monkey Cloning. Cell Reprogram 2024; 26:120-123. [PMID: 39088354 DOI: 10.1089/cell.2024.0048] [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: 08/03/2024] Open
Abstract
Cloning by somatic cell nuclear transfer (SCNT) remained challenging for Rhesus monkeys, mostly due to its low efficiency and neonatal death. Genome-scale analyses revealed that monkey SCNT embryos displayed widespread DNA methylation and transcriptional alterations, thus including loss of genomic imprinting that correlated with placental dysfunction. The transfer of inner cell masses (ICM) from cloned blastocysts into ICM-depleted fertilized embryos rescued placental insufficiency and gave rise to a cloned Rhesus monkey that reached adulthood without noticeable abnormalities.
Collapse
Affiliation(s)
- Marcelo Tigre Moura
- Departamento de Biologia Celular e Molecular, Centro de Biotecnologia, Universidade Federal da Paraíba-UFPB, João Pessoa, Brazil
| |
Collapse
|
30
|
Liberali P, Schier AF. The evolution of developmental biology through conceptual and technological revolutions. Cell 2024; 187:3461-3495. [PMID: 38906136 DOI: 10.1016/j.cell.2024.05.053] [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: 04/12/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/23/2024]
Abstract
Developmental biology-the study of the processes by which cells, tissues, and organisms develop and change over time-has entered a new golden age. After the molecular genetics revolution in the 80s and 90s and the diversification of the field in the early 21st century, we have entered a phase when powerful technologies provide new approaches and open unexplored avenues. Progress in the field has been accelerated by advances in genomics, imaging, engineering, and computational biology and by emerging model systems ranging from tardigrades to organoids. We summarize how revolutionary technologies have led to remarkable progress in understanding animal development. We describe how classic questions in gene regulation, pattern formation, morphogenesis, organogenesis, and stem cell biology are being revisited. We discuss the connections of development with evolution, self-organization, metabolism, time, and ecology. We speculate how developmental biology might evolve in an era of synthetic biology, artificial intelligence, and human engineering.
Collapse
Affiliation(s)
- Prisca Liberali
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland; University of Basel, Basel, Switzerland.
| | | |
Collapse
|
31
|
Gui L, Zhong Q, Yang J, Sun J, Lu J, Picton HM, Li C. Acquisition of 2C-like totipotency through defined maternal-effect factors. Stem Cells 2024; 42:581-592. [PMID: 38655883 DOI: 10.1093/stmcls/sxae029] [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: 08/16/2023] [Accepted: 03/25/2024] [Indexed: 04/26/2024]
Abstract
Fully grown oocytes have the natural ability to transform 2 terminally differentiated gametes into a totipotent zygote representing the acquisition of totipotency. This process wholly depends on maternal-effect factors (MFs). MFs stored in the eggs are therefore likely to be able to induce cellular reprogramming to a totipotency state. Here we report the generation of totipotent-like stem cells from mESCs using 4MFs Hsf1, Zar1, Padi6, and Npm2, designated as MFiTLSCs. MFiTLSCs exhibited a unique and inherent capability to differentiate into embryonic and extraembryonic derivatives. Transcriptomic analysis revealed that MFiTLSCs are enriched with 2-cell-specific genes that appear to synergistically induce a transcriptional repressive state, in that parental genomes are remodeled to a poised transcriptional repression state while totipotency is established following fertilization. This method to derive MFiTLSCs could help advance the understanding of fate determinations of totipotent stem cells in a physiological context and establish a foundation for the development of oocyte biology-based reprogramming technology.
Collapse
Affiliation(s)
- Liming Gui
- Institute of Obstetrics and Gynaecology, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong Province 518036, People's Republic of China
- Department of Obstetrics and Gynaecology, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province 518036, People's Republic of China
- Centre for Tissue Engineering and Stem Cell Research, Guizhou Medical University, Guiyang, Guizhou Province 550004, People's Republic of China
| | - Qin Zhong
- Centre for Tissue Engineering and Stem Cell Research, Guizhou Medical University, Guiyang, Guizhou Province 550004, People's Republic of China
| | - Jue Yang
- Centre for Tissue Engineering and Stem Cell Research, Guizhou Medical University, Guiyang, Guizhou Province 550004, People's Republic of China
| | - Jiajia Sun
- Institute of Obstetrics and Gynaecology, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong Province 518036, People's Republic of China
| | - Jianping Lu
- Reproduction and Early Development Research Group, Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Helen M Picton
- Reproduction and Early Development Research Group, Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Changzhong Li
- Institute of Obstetrics and Gynaecology, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong Province 518036, People's Republic of China
- Department of Obstetrics and Gynaecology, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province 518036, People's Republic of China
| |
Collapse
|
32
|
Peterson L, Yacoub MH, Ayares D, Yamada K, Eisenson D, Griffith BP, Mohiuddin MM, Eyestone W, Venter JC, Smolenski RT, Rothblatt M. Physiological basis for xenotransplantation from genetically modified pigs to humans. Physiol Rev 2024; 104:1409-1459. [PMID: 38517040 PMCID: PMC11390123 DOI: 10.1152/physrev.00041.2023] [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: 10/26/2023] [Revised: 03/06/2024] [Accepted: 03/14/2024] [Indexed: 03/23/2024] Open
Abstract
The collective efforts of scientists over multiple decades have led to advancements in molecular and cellular biology-based technologies including genetic engineering and animal cloning that are now being harnessed to enhance the suitability of pig organs for xenotransplantation into humans. Using organs sourced from pigs with multiple gene deletions and human transgene insertions, investigators have overcome formidable immunological and physiological barriers in pig-to-nonhuman primate (NHP) xenotransplantation and achieved prolonged pig xenograft survival. These studies informed the design of Revivicor's (Revivicor Inc, Blacksburg, VA) genetically engineered pigs with 10 genetic modifications (10 GE) (including the inactivation of 4 endogenous porcine genes and insertion of 6 human transgenes), whose hearts and kidneys have now been studied in preclinical human xenotransplantation models with brain-dead recipients. Additionally, the first two clinical cases of pig-to-human heart xenotransplantation were recently performed with hearts from this 10 GE pig at the University of Maryland. Although this review focuses on xenotransplantation of hearts and kidneys, multiple organs, tissues, and cell types from genetically engineered pigs will provide much-needed therapeutic interventions in the future.
Collapse
Affiliation(s)
- Leigh Peterson
- United Therapeutics Corporation, Silver Spring, Maryland, United States
| | | | - David Ayares
- United Therapeutics Corporation, Silver Spring, Maryland, United States
| | - Kazuhiko Yamada
- Department of Surgery, Division of Transplantation, Johns Hopkins Medicine, Baltimore, Maryland, United States
| | - Daniel Eisenson
- Department of Surgery, Division of Transplantation, Johns Hopkins Medicine, Baltimore, Maryland, United States
| | - Bartley P Griffith
- University of Maryland Medical Center, Baltimore, Maryland, United States
| | | | - Willard Eyestone
- United Therapeutics Corporation, Silver Spring, Maryland, United States
| | - J Craig Venter
- J. Craig Venter Institute, Rockville, Maryland, United States
| | | | - Martine Rothblatt
- United Therapeutics Corporation, Silver Spring, Maryland, United States
| |
Collapse
|
33
|
Yoneyama Y, Zhang RR, Kimura M, Cai Y, Adam M, Parameswaran S, Masaki H, Mizuno N, Bhadury J, Maezawa S, Ochiai H, Nakauchi H, Potter SS, Weirauch MT, Takebe T. Inter-cellular mRNA Transfer Alters Human Pluripotent Stem Cell State. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.600209. [PMID: 38979277 PMCID: PMC11230441 DOI: 10.1101/2024.06.27.600209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Inter-cellular transmission of mRNA is being explored in mammalian species using immortal cell lines (1-3). Here, we uncover an inter-cellular mRNA transfer phenomenon that allows for the adaptation and reprogramming of human primed pluripotent stem cells (hPSCs). This process is induced by the direct cell contact-mediated coculture with mouse embryonic stem cells (mESCs) under the condition impermissible for human primed PSC culture. Mouse-derived mRNA contents are transmitted into adapted hPSCs only in the coculture. Transfer-specific mRNA analysis show the enrichment for divergent biological pathways involving transcription/translational machinery and stress-coping mechanisms, wherein such transfer is diminished when direct cell contacts are lost. After 5 days of mESC culture, surface marker analysis, and global gene profiling confirmed that mRNA transfer-prone hPSC efficiently gains a naïve-like state. Furthermore, transfer-specific knockdown experiments targeting mouse-specific transcription factor-coding mRNAs in hPSC show that mouse-derived Tfcp2l1, Tfap2c, and Klf4 are indispensable for human naïve-like conversion. Thus, inter-species mRNA transfer triggers cellular reprogramming in mammalian cells. Our results support that episodic mRNA transfer can occur in cell cooperative and competitive processes(4), which provides a fresh perspective on understanding the roles of mRNA mobility for intra- and inter-species cellular communications.
Collapse
Affiliation(s)
- Yosuke Yoneyama
- Institute of Research, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
- Department of Genome Biology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Ran-Ran Zhang
- Divisions of Gastroenterology, Hepatology & Nutrition, Developmental Biology and Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
| | - Masaki Kimura
- Divisions of Gastroenterology, Hepatology & Nutrition, Developmental Biology and Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
| | - Yuqi Cai
- Divisions of Gastroenterology, Hepatology & Nutrition, Developmental Biology and Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
| | - Mike Adam
- Divisions of Gastroenterology, Hepatology & Nutrition, Developmental Biology and Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
| | - Sreeja Parameswaran
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
| | - Hideki Masaki
- Division of Stem Cell Therapy, Institute of Medical Science, University of Tokyo, Tokyo, 108-8639, Japan
| | - Naoaki Mizuno
- Division of Stem Cell Therapy, Institute of Medical Science, University of Tokyo, Tokyo, 108-8639, Japan
| | - Joydeep Bhadury
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - So Maezawa
- Faculty of Science and Technology, Department of Applied Biological Science, Tokyo University of Science, Chiba, 278-8510, Japan
| | - Hiroshi Ochiai
- Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-0054, Japan
| | - Hiromitsu Nakauchi
- Division of Stem Cell Therapy, Institute of Medical Science, University of Tokyo, Tokyo, 108-8639, Japan
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA 94305, USA
| | - S. Steven Potter
- Divisions of Gastroenterology, Hepatology & Nutrition, Developmental Biology and Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
| | - Matthew T. Weirauch
- Divisions of Gastroenterology, Hepatology & Nutrition, Developmental Biology and Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
- Center for Autoimmune Genomics and Etiology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
| | - Takanori Takebe
- Institute of Research, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
- Department of Genome Biology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
- Divisions of Gastroenterology, Hepatology & Nutrition, Developmental Biology and Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
- Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
34
|
Ledesma AV, Van Eenennaam AL. Global status of gene edited animals for agricultural applications. Vet J 2024; 305:106142. [PMID: 38788996 DOI: 10.1016/j.tvjl.2024.106142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 05/21/2024] [Accepted: 05/21/2024] [Indexed: 05/26/2024]
Abstract
Gene editing (GnEd) involves using a site-directed nuclease to introduce a double-strand break (DSB) at a targeted location in the genome. A literature search was performed on the use of GnEd in animals for agricultural applications. Data was extracted from 212 peer-reviewed articles that described the production of at least one living animal employing GnEd technologies for agricultural purposes. The most common GnEd system reported was CRISPR/Cas9, and the most frequent type of edit was the unguided insertion or deletion resulting from the repair of the targeted DSB leading to a knock-out (KO) mutation. Animal groups included in the reviewed papers were ruminants (cattle, sheep, goats, n=63); monogastrics (pigs and rabbits, n=60); avian (chicken, duck, quail, n=17); aquatic (many species, n=65), and insects (honeybee, silkworm, n=7). Yield (32%), followed by reproduction (21%) and disease resistance (17%) were the most commonly targeted traits. Over half of the reviewed papers had Chinese first-authorship. Several countries, including Argentina, Australia, Brazil, Colombia and Japan, have adopted a regulatory policy that considers KO mutations introduced following GnEd DSB repair as akin to natural genetic variation, and therefore treat these GnEd animals analogously to those produced using conventional breeding. This approach has resulted in a non-GMO determination for a small number of GnEd food animal applications, including three species of GnEd KO fast-growing fish, (red sea bream, olive flounder and tiger pufferfish in Japan), KO fish and cattle in Argentina and Brazil, and porcine reproductive and respiratory syndrome (PRRS) virus disease-resistant KO pigs in Colombia.
Collapse
Affiliation(s)
- Alba V Ledesma
- Department of Animal Science, University of California, Davis, CA 95616, USA
| | | |
Collapse
|
35
|
Ji K, Park K, Kim D, Kim E, Kil T, Kim M. Accomplishment of canine cloning through in vitro matured oocytes: a pioneering milestone. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2024; 66:577-586. [PMID: 38975582 PMCID: PMC11222123 DOI: 10.5187/jast.2024.e18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 07/09/2024]
Abstract
The in vitro maturation (IVM) rate of canine oocytes remains low compared to other mammals due to their unique reproductive characteristics. This study aimed to explore the effect of hormone supplementation during the IVM of canine immature oocytes on nuclear maturation and subsequently assess its potential application in canine somatic cell nuclear transfer (SCNT). Immature oocytes were collected and cultured in an IVM medium supplemented with hormones (follicle-stimulating hormone [FSH] and progesterone [P4]) or without hormones (control) for 24 hours. The maturation rates of oocytes in the hormone-treated group (94.92 ± 3.15%) were significantly higher than those in the control group (61.01 ± 4.23%). Both in vitro and in vivo matured oocytes underwent NT to evaluate their utility, and the fusion rates were higher in the in vitro matured group than those in the vivo matured group, not significant between in vivo and in vitro matured group (73.28% and 82.35%, respectively). As a result, 14 fused embryos from the in vitro matured group were transferred into two surrogates, with one surrogate achieving a successful pregnancy and delivering four puppies. Whereas in the in vivo matured group, 85 fused embryos were transferred to 8 surrogate mothers, leading to three surrogates becoming pregnant and delivering one, four, and two puppies. The pregnancy rates were not significant between both groups (50% and 37.50%), but the number of offspring exhibited a significant difference (28.57% and 8.23%). In conclusion, we achieved a remarkable milestone by successfully producing cloned puppies using in vitro matured oocytes, underscoring the feasibility of canine cloning from in vitro recovered oocytes. It is important to note that this study focused only on immature oocytes after ovulation and only during the estrus stage. Further research targeting other stages of the estrous cycle could potentially enhance canine cloning efficiency.
Collapse
Affiliation(s)
- Kukbin Ji
- Division of Animal and Dairy Science, College of Agriculture and Life Science, Chungnam National University, Daejeon 34134, Korea
| | | | - Dongern Kim
- Division of Animal and Dairy Science, College of Agriculture and Life Science, Chungnam National University, Daejeon 34134, Korea
| | | | - Taeyoung Kil
- Department of Social Welfare, Joongbu University, Geumsan 32713, Korea
| | - Minkyu Kim
- Division of Animal and Dairy Science, College of Agriculture and Life Science, Chungnam National University, Daejeon 34134, Korea
- MK Biotech Co., LTD., Daejeon 34134, Korea
| |
Collapse
|
36
|
Zhang M, Zhai Y, An X, Li Q, Zhang D, Zhou Y, Zhang S, Dai X, Li Z. DNA methylation regulates RNA m 6A modification through transcription factor SP1 during the development of porcine somatic cell nuclear transfer embryos. Cell Prolif 2024; 57:e13581. [PMID: 38095020 PMCID: PMC11056710 DOI: 10.1111/cpr.13581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/12/2023] [Accepted: 11/15/2023] [Indexed: 01/12/2024] Open
Abstract
Epigenetic modifications play critical roles during somatic cell nuclear transfer (SCNT) embryo development. Whether RNA N6-methyladenosine (m6A) affects the developmental competency of SCNT embryos remains unclear. Here, we showed that porcine bone marrow mesenchymal stem cells (pBMSCs) presented higher RNA m6A levels than those of porcine embryonic fibroblasts (pEFs). SCNT embryos derived from pBMSCs had higher RNA m6A levels, cleavage, and blastocyst rates than those from pEFs. Compared with pEFs, the promoter region of METTL14 presented a hypomethylation status in pBMSCs. Mechanistically, DNA methylation regulated METTL14 expression by affecting the accessibility of transcription factor SP1 binding, highlighting the role of the DNA methylation/SP1/METTL14 pathway in donor cells. Inhibiting the DNA methylation level in donor cells increased the RNA m6A level and improved the development efficiency of SCNT embryos. Overexpression of METTL14 significantly increased the RNA m6A level in donor cells and the development efficiency of SCNT embryos, whereas knockdown of METTL14 suggested the opposite result. Moreover, we revealed that RNA m6A-regulated TOP2B mRNA stability, translation level, and DNA damage during SCNT embryo development. Collectively, our results highlight the crosstalk between RNA m6A and DNA methylation, and the crucial role of RNA m6A during nuclear reprogramming in SCNT embryo development.
Collapse
Affiliation(s)
- Meng Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of EducationThe First Hospital of Jilin UniversityChangchunJilinChina
| | - Yanhui Zhai
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of EducationThe First Hospital of Jilin UniversityChangchunJilinChina
| | - Xinglan An
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of EducationThe First Hospital of Jilin UniversityChangchunJilinChina
| | - Qi Li
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of EducationThe First Hospital of Jilin UniversityChangchunJilinChina
| | - Daoyu Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of EducationThe First Hospital of Jilin UniversityChangchunJilinChina
| | - Yongfeng Zhou
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of EducationThe First Hospital of Jilin UniversityChangchunJilinChina
| | - Sheng Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of EducationThe First Hospital of Jilin UniversityChangchunJilinChina
| | - Xiangpeng Dai
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of EducationThe First Hospital of Jilin UniversityChangchunJilinChina
| | - Ziyi Li
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of EducationThe First Hospital of Jilin UniversityChangchunJilinChina
| |
Collapse
|
37
|
Chen Y, Ye Z, Lin M, Zhu L, Xu L, Wang X. Deciphering the Epigenetic Landscape: Placental Development and Its Role in Pregnancy Outcomes. Stem Cell Rev Rep 2024; 20:996-1014. [PMID: 38457061 DOI: 10.1007/s12015-024-10699-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2024] [Indexed: 03/09/2024]
Abstract
The placenta stands out as a unique, transitory, and multifaceted organ, essential to the optimal growth and maturation of the fetus. Functioning as a vital nexus between the maternal and fetal circulatory systems, it oversees the critical exchange of nutrients and waste. This exchange is facilitated by placental cells, known as trophoblasts, which adeptly invade and remodel uterine blood vessels. Deviations in placental development underpin a slew of pregnancy complications, notably fetal growth restriction (FGR), preeclampsia (PE), recurrent spontaneous abortions (RSA), and preterm birth. Central to placental function and development is epigenetic regulation. Despite its importance, the intricate mechanisms by which epigenetics influence the placenta are not entirely elucidated. Recently, the scientific community has turned its focus to parsing out the epigenetic alterations during placental development, such as variations in promoter DNA methylation, genomic imprints, and shifts in non-coding RNA expression. By establishing correlations between epigenetic shifts in the placenta and pregnancy complications, researchers are unearthing invaluable insights into the biology and pathophysiology of these conditions. This review seeks to synthesize the latest findings on placental epigenetic regulation, spotlighting its crucial role in shaping fetal growth trajectories and development. Through this lens, we underscore the overarching significance of the placenta in the larger narrative of gestational health.
Collapse
Affiliation(s)
- Yujia Chen
- Medical Research Center, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- National Health Commission (NHC), Key Laboratory of Technical Evaluation of Fertility Regulation for Non-Human Primate, Fujian Maternity and Child Health Hospital, Fuzhou, China
| | - Zhoujie Ye
- Medical Research Center, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- National Health Commission (NHC), Key Laboratory of Technical Evaluation of Fertility Regulation for Non-Human Primate, Fujian Maternity and Child Health Hospital, Fuzhou, China
| | - Meijia Lin
- Department of Pathology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| | - Liping Zhu
- Medical Research Center, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- National Health Commission (NHC), Key Laboratory of Technical Evaluation of Fertility Regulation for Non-Human Primate, Fujian Maternity and Child Health Hospital, Fuzhou, China
| | - Liangpu Xu
- Medical Genetic Diagnosis and Therapy Center of Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Provincial Key Laboratory of Prenatal Diagnosis and Birth Defect, Fuzhou, China.
| | - Xinrui Wang
- Medical Research Center, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China.
- National Health Commission (NHC), Key Laboratory of Technical Evaluation of Fertility Regulation for Non-Human Primate, Fujian Maternity and Child Health Hospital, Fuzhou, China.
| |
Collapse
|
38
|
Mariano CG, de Oliveira VC, Ambrósio CE. Gene editing in small and large animals for translational medicine: a review. Anim Reprod 2024; 21:e20230089. [PMID: 38628493 PMCID: PMC11019828 DOI: 10.1590/1984-3143-ar2023-0089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 02/16/2024] [Indexed: 04/19/2024] Open
Abstract
The CRISPR/Cas9 system is a simpler and more versatile method compared to other engineered nucleases such as Zinc Finger Nucleases (ZFNs) and Transcription Activator-Like Effector Nucleases (TALENs), and since its discovery, the efficiency of CRISPR-based genome editing has increased to the point that multiple and different types of edits can be made simultaneously. These advances in gene editing have revolutionized biotechnology by enabling precise genome editing with greater simplicity and efficacy than ever before. This tool has been successfully applied to a wide range of animal species, including cattle, pigs, dogs, and other small animals. Engineered nucleases cut the genome at specific target positions, triggering the cell's mechanisms to repair the damage and introduce a mutation to a specific genomic site. This review discusses novel genome-based CRISPR/Cas9 editing tools, methods developed to improve efficiency and specificity, the use of gene-editing on animal models and translational medicine, and the main challenges and limitations of CRISPR-based gene-editing approaches.
Collapse
Affiliation(s)
- Clésio Gomes Mariano
- Departamento de Medicina Veterinária, Faculdade de Zootecnia e Engenharia de Alimentos, Universidade de São Paulo – USP, Pirassununga, SP, Brasil
| | - Vanessa Cristina de Oliveira
- Departamento de Medicina Veterinária, Faculdade de Zootecnia e Engenharia de Alimentos, Universidade de São Paulo – USP, Pirassununga, SP, Brasil
| | - Carlos Eduardo Ambrósio
- Departamento de Medicina Veterinária, Faculdade de Zootecnia e Engenharia de Alimentos, Universidade de São Paulo – USP, Pirassununga, SP, Brasil
| |
Collapse
|
39
|
Kang JS, Joo MD, Lee SH, Kang SM, Haider Z, Perera CD, Idrees M, Jin Y, Kong IK. Effect of additional cytoplasm injection on the cloned bovine embryo organelle distribution and stress mitigation. Theriogenology 2024; 216:12-19. [PMID: 38147714 DOI: 10.1016/j.theriogenology.2023.11.031] [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: 07/27/2023] [Revised: 11/29/2023] [Accepted: 11/29/2023] [Indexed: 12/28/2023]
Abstract
Although somatic cell nuclear transfer (SCNT) is a critical component of animal cloning, this approach has several issues. We previously introduced the cytoplasm injection cloning technology (CICT), which significantly improves the quality and quantity of cloned embryos. This study examined the residual status of fused cytoplasmic organelles, such as the endoplasmic reticulum (ER) and lysosomes, in the CICT group during early embryo development. We found that extra-cytoplasmic organelles stained using the ER-Tracker™ Green dye and LysoTracker™ Deep Red probe were fused and dispersed throughout the recipient oocyte and were still visible in day 8 blastocysts. We screened for ER stress, autophagy, and apoptosis-related genes to elucidate the association between the added organelles and improved embryo quality in CICT-cloned embryos. We found that CHOP, ATF4, ATG5, ATG7, and LC3 genes showed non-significantly up- or downregulated expression between CICT- and in vitro fertilization (IVF)-derived embryos but showed significantly (p < 0.05) upregulated expression in SCNT-cloned embryos. Surprisingly, a non-significant difference in the expression of some genes, such as ATF6 and caspase-3, was observed between the CICT- and SCNT-cloned embryos. Our findings imply that compared to conventional SCNT cloning, CICT-derived cloned embryos with additional cytoplasm have much higher organelle activity, lower autophagy, lower rates of apoptosis, and higher embryo development rates.
Collapse
Affiliation(s)
- Ji-Su Kang
- Division of Applied Life Science (BK21 Four), Graduate School of Applied Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea.
| | - Myeong-Don Joo
- Division of Applied Life Science (BK21 Four), Graduate School of Applied Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea.
| | - Seo-Hyeon Lee
- Division of Applied Life Science (BK21 Four), Graduate School of Applied Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Seon-Min Kang
- Division of Applied Life Science (BK21 Four), Graduate School of Applied Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Zaheer Haider
- Division of Applied Life Science (BK21 Four), Graduate School of Applied Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Chalani Dilshani Perera
- Division of Applied Life Science (BK21 Four), Graduate School of Applied Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Muhammad Idrees
- Division of Applied Life Science (BK21 Four), Graduate School of Applied Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea; Institute of Agriculture and Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Yongxun Jin
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, 130062, Jilin, PR China.
| | - Il-Keun Kong
- Department of Laboratory Animals, Jilin Provincial Key Laboratory of Animal Model, Jilin University, Changchun, 130062, Jilin, PR China; Division of Applied Life Science (BK21 Four), Graduate School of Applied Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea; Institute of Agriculture and Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea; Thekingkong Co. Ltd., Gyeongsang National University, Jinju, 52828, Republic of Korea.
| |
Collapse
|
40
|
Beghini DG, Kasai-Brunswick TH, Henriques-Pons A. Induced Pluripotent Stem Cells in Drug Discovery and Neurodegenerative Disease Modelling. Int J Mol Sci 2024; 25:2392. [PMID: 38397069 PMCID: PMC10889263 DOI: 10.3390/ijms25042392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 02/25/2024] Open
Abstract
Induced pluripotent stem cells (iPSCs) are derived from reprogrammed adult somatic cells. These adult cells are manipulated in vitro to express genes and factors essential for acquiring and maintaining embryonic stem cell (ESC) properties. This technology is widely applied in many fields, and much attention has been given to developing iPSC-based disease models to validate drug discovery platforms and study the pathophysiological molecular processes underlying disease onset. Especially in neurological diseases, there is a great need for iPSC-based technological research, as these cells can be obtained from each patient and carry the individual's bulk of genetic mutations and unique properties. Moreover, iPSCs can differentiate into multiple cell types. These are essential characteristics, since the study of neurological diseases is affected by the limited access to injury sites, the need for in vitro models composed of various cell types, the complexity of reproducing the brain's anatomy, the challenges of postmortem cell culture, and ethical issues. Neurodegenerative diseases strongly impact global health due to their high incidence, symptom severity, and lack of effective therapies. Recently, analyses using disease specific, iPSC-based models confirmed the efficacy of these models for testing multiple drugs. This review summarizes the advances in iPSC technology used in disease modelling and drug testing, with a primary focus on neurodegenerative diseases, including Parkinson's and Alzheimer's diseases.
Collapse
Affiliation(s)
- Daniela Gois Beghini
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, RJ, Brazil;
| | - Tais Hanae Kasai-Brunswick
- Centro Nacional de Biologia Estrutural e Bioimagem, CENABIO, Universidade Federal do Rio de Janeiro, Seropédica 23890-000, RJ, Brazil;
- Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, INCT-REGENERA, Universidade Federal do Rio de Janeiro, Seropédica 23890-000, RJ, Brazil
| | - Andrea Henriques-Pons
- Laboratório de Inovações em Terapias, Ensino e Bioprodutos, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, RJ, Brazil;
| |
Collapse
|
41
|
Cowl VB, Comizzoli P, Appeltant R, Bolton RL, Browne RK, Holt WV, Penfold LM, Swegen A, Walker SL, Williams SA. Cloning for the Twenty-First Century and Its Place in Endangered Species Conservation. Annu Rev Anim Biosci 2024; 12:91-112. [PMID: 37988633 DOI: 10.1146/annurev-animal-071423-093523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2023]
Abstract
Cloning as it relates to the animal kingdom generally refers to the production of genetically identical individuals. Because cloning is increasingly the subject of renewed attention as a tool for rescuing endangered or extinct species, it seems timely to dissect the role of the numerous reproductive techniques encompassed by this term in animal species conservation. Although cloning is typically associated with somatic cell nuclear transfer, the recent advent of additional techniques that allow genome replication without genetic recombination demands that the use of induced pluripotent stem cells to generate gametes or embryos, as well as older methods such as embryo splitting, all be included in this discussion. Additionally, the phenomenon of natural cloning (e.g., a subset of fish, birds, invertebrates, and reptilian species that reproduce via parthenogenesis) must also be pointed out. Beyond the biology of these techniques are practical considerations and the ethics of using cloning and associated procedures in endangered or extinct species. All of these must be examined in concert to determine whether cloning has a place in species conservation. Therefore, we synthesize progress in cloning and associated techniques and dissect the practical and ethical aspects of these methods as they pertain to endangered species conservation.
Collapse
Affiliation(s)
- Veronica B Cowl
- North of England Zoological Society (Chester Zoo), Chester, United Kingdom;
- European Association of Zoos and Aquaria, Amsterdam, The Netherlands
| | - Pierre Comizzoli
- Smithsonian's National Zoo and Conservation Biology Institute, Washington, DC, USA;
| | - Ruth Appeltant
- Gamete Research Centre, Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium;
| | | | - Robert K Browne
- Sustainability America, Sarteneja, Corozal District, Belize;
| | - William V Holt
- Department of Oncology and Metabolism, The Medical School, University of Sheffield, Sheffield, United Kingdom;
| | - Linda M Penfold
- South East Zoo Alliance for Reproduction & Conservation, Yulee, Florida, USA;
| | - Aleona Swegen
- Priority Research Centre for Reproductive Science, University of Newcastle, Callaghan, New South Wales, Australia;
| | - Susan L Walker
- North of England Zoological Society (Chester Zoo), Chester, United Kingdom;
- Nature's SAFE, Whitchurch, Shropshire, United Kingdom;
| | - Suzannah A Williams
- Nature's SAFE, Whitchurch, Shropshire, United Kingdom;
- Nuffield Department of Women's and Reproductive Health, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom;
| |
Collapse
|
42
|
Paine PT, Nguyen A, Ocampo A. Partial cellular reprogramming: A deep dive into an emerging rejuvenation technology. Aging Cell 2024; 23:e14039. [PMID: 38040663 PMCID: PMC10861195 DOI: 10.1111/acel.14039] [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: 06/06/2023] [Revised: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 12/03/2023] Open
Abstract
Aging and age-associated disease are a major medical and societal burden in need of effective treatments. Cellular reprogramming is a biological process capable of modulating cell fate and cellular age. Harnessing the rejuvenating benefits without altering cell identity via partial cellular reprogramming has emerged as a novel translational strategy with therapeutic potential and strong commercial interests. Here, we explore the aging-related benefits of partial cellular reprogramming while examining limitations and future directions for the field.
Collapse
Affiliation(s)
- Patrick T. Paine
- Department of Biomedical Sciences, Faculty of Biology and MedicineUniversity of LausanneLausanneVaudSwitzerland
- Center for Virology and Vaccine ResearchHarvard Medical SchoolBostonMassachusettsUSA
- Present address:
McGovern Institute for Brain Research at MIT, Massachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | | | - Alejandro Ocampo
- Department of Biomedical Sciences, Faculty of Biology and MedicineUniversity of LausanneLausanneVaudSwitzerland
- EPITERNA SAEpalingesSwitzerland
| |
Collapse
|
43
|
Ward MA, Roberts RM, Ward WS. Ryuzo Yanagimachi: Pioneer in fertilization and assisted reproductive biology technology. Proc Natl Acad Sci U S A 2024; 121:e2320501121. [PMID: 38190518 PMCID: PMC10801875 DOI: 10.1073/pnas.2320501121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2024] Open
Affiliation(s)
- Monika A. Ward
- Department of Anatomy, Biochemistry, and Physiology, Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI96822
| | - R. Michael Roberts
- University of Missouri-Columbia - Animal Sciences, 240B Bond Life Sciences Center, Columbia, MO65211
| | - W. Steven Ward
- Department of Anatomy, Biochemistry, and Physiology, Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI96822
- Department of Obstetrics and Gynecology and Women’s Health, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI96822
| |
Collapse
|
44
|
Liao Z, Zhang J, Sun S, Li Y, Xu Y, Li C, Cao J, Nie Y, Niu Z, Liu J, Lu F, Liu Z, Sun Q. Reprogramming mechanism dissection and trophoblast replacement application in monkey somatic cell nuclear transfer. Nat Commun 2024; 15:5. [PMID: 38228612 PMCID: PMC10791636 DOI: 10.1038/s41467-023-43985-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 11/27/2023] [Indexed: 01/18/2024] Open
Abstract
Somatic cell nuclear transfer (SCNT) successfully clones cynomolgus monkeys, but the efficiency remains low due to a limited understanding of the reprogramming mechanism. Notably, no rhesus monkey has been cloned through SCNT so far. Our study conducts a comparative analysis of multi-omics datasets, comparing embryos resulting from intracytoplasmic sperm injection (ICSI) with those from SCNT. Our findings reveal a widespread decrease in DNA methylation and the loss of imprinting in maternally imprinted genes within SCNT monkey blastocysts. This loss of imprinting persists in SCNT embryos cultured in-vitro until E17 and in full-term SCNT placentas. Additionally, histological examination of SCNT placentas shows noticeable hyperplasia and calcification. To address these defects, we develop a trophoblast replacement method, ultimately leading to the successful cloning of a healthy male rhesus monkey. These discoveries provide valuable insights into the reprogramming mechanism of monkey SCNT and introduce a promising strategy for primate cloning.
Collapse
Affiliation(s)
- Zhaodi Liao
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, 200031, China
- Shanghai Center for Brain Science and Brain-Inspired Technology, Shanghai, 201210, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jixiang Zhang
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Shiyu Sun
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, 200031, China
- Shanghai Center for Brain Science and Brain-Inspired Technology, Shanghai, 201210, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuzhuo Li
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, 200031, China
- Shanghai Center for Brain Science and Brain-Inspired Technology, Shanghai, 201210, China
| | - Yuting Xu
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, 200031, China
- Shanghai Center for Brain Science and Brain-Inspired Technology, Shanghai, 201210, China
| | - Chunyang Li
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, 200031, China
- Shanghai Center for Brain Science and Brain-Inspired Technology, Shanghai, 201210, China
| | - Jing Cao
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, 200031, China
- Shanghai Center for Brain Science and Brain-Inspired Technology, Shanghai, 201210, China
| | - Yanhong Nie
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, 200031, China
- Shanghai Center for Brain Science and Brain-Inspired Technology, Shanghai, 201210, China
| | - Zhuoyue Niu
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jingwen Liu
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Falong Lu
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Zhen Liu
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, 200031, China.
- Shanghai Center for Brain Science and Brain-Inspired Technology, Shanghai, 201210, China.
| | - Qiang Sun
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, 200031, China.
- Shanghai Center for Brain Science and Brain-Inspired Technology, Shanghai, 201210, China.
| |
Collapse
|
45
|
Kwon DH, Gim GM, Yum SY, Jang G. Current status and future of gene engineering in livestock. BMB Rep 2024; 57:50-59. [PMID: 38053297 PMCID: PMC10828428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/23/2023] [Accepted: 12/04/2023] [Indexed: 12/07/2023] Open
Abstract
The application of gene engineering in livestock is necessary for various reasons, such as increasing productivity and producing disease resistance and biomedicine models. Overall, gene engineering provides benefits to the agricultural and research aspects, and humans. In particular, productivity can be increased by producing livestock with enhanced growth and improved feed conversion efficiency. In addition, the application of the disease resistance models prevents the spread of infectious diseases, which reduces the need for treatment, such as the use of antibiotics; consequently, it promotes the overall health of the herd and reduces unexpected economic losses. The application of biomedicine could be a valuable tool for understanding specific livestock diseases and improving human welfare through the development and testing of new vaccines, research on human physiology, such as human metabolism or immune response, and research and development of xenotransplantation models. Gene engineering technology has been evolving, from random, time-consuming, and laborious methods to specific, time-saving, convenient, and stable methods. This paper reviews the overall trend of genetic engineering technologies development and their application for efficient production of genetically engineered livestock, and provides examples of technologies approved by the United States (US) Food and Drug Administration (FDA) for application in humans. [BMB Reports 2024; 57(1): 50-59].
Collapse
Affiliation(s)
- Dong-Hyeok Kwon
- Laboratory of Theriogenology, College of Veterinary Medicine, Research Institute for Veterinary Science, BK21 FOUR Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul 08826, Korea
| | | | | | - Goo Jang
- Laboratory of Theriogenology, College of Veterinary Medicine, Research Institute for Veterinary Science, BK21 FOUR Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul 08826, Korea
- LARTBio Inc., Gwangmyeong 14322, Korea
| |
Collapse
|
46
|
Darban YM, Askari H, Ghasemi-Kasman M, Yavarpour-Bali H, Dehpanah A, Gholizade P, Nosratiyan N. The Role of Induced Pluripotent Stem Cells in the Treatment of Stroke. Curr Neuropharmacol 2024; 22:2368-2383. [PMID: 39403058 PMCID: PMC11451314 DOI: 10.2174/1570159x22666240603084558] [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: 10/01/2023] [Revised: 01/14/2024] [Accepted: 01/16/2024] [Indexed: 10/19/2024] Open
Abstract
Stroke is a neurological disorder with high disability and mortality rates. Almost 80% of stroke cases are ischemic stroke, and the remaining are hemorrhagic stroke. The only approved treatment for ischemic stroke is thrombolysis and/or thrombectomy. However, these treatments cannot sufficiently relieve the disease outcome, and many patients remain disabled even after effective thrombolysis. Therefore, rehabilitative therapies are necessary to induce remodeling in the brain. Currently, stem cell transplantation, especially via the use of induced pluripotent stem cells (iPSCs), is considered a promising alternative therapy for stimulating neurogenesis and brain remodeling. iPSCs are generated from somatic cells by specific transcription factors. The biological functions of iPSCs are similar to those of embryonic stem cells (ESCs), including immunomodulation, reduced cerebral blood flow, cerebral edema, and autophagy. Although iPSC therapy plays a promising role in both hemorrhagic and ischemic stroke, its application is associated with certain limitations. Tumor formation, immune rejection, stem cell survival, and migration are some concerns associated with stem cell therapy. Therefore, cell-free therapy as an alternative method can overcome these limitations. This study reviews the therapeutic application of iPSCs in stroke models and the underlying mechanisms and constraints of these cells. Moreover, cell-free therapy using exosomes, apoptotic bodies, and microvesicles as alternative treatments is discussed.
Collapse
Affiliation(s)
| | - Hamid Askari
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Maryam Ghasemi-Kasman
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
- Department of Physiology, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | | | - Amirabbas Dehpanah
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Parnia Gholizade
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Nasrin Nosratiyan
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
47
|
Schalich KM, Koganti PP, Castillo JM, Reiff OM, Cheong SH, Selvaraj V. The uterine secretory cycle: recurring physiology of endometrial outputs that setup the uterine luminal microenvironment. Physiol Genomics 2024; 56:74-97. [PMID: 37694291 DOI: 10.1152/physiolgenomics.00035.2023] [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: 05/03/2023] [Revised: 09/08/2023] [Accepted: 09/08/2023] [Indexed: 09/12/2023] Open
Abstract
Conserved in female reproduction across all mammalian species is the estrous cycle and its regulation by the hypothalamic-pituitary-gonadal (HPG) axis, a collective of intersected hormonal events that are crucial for ensuring uterine fertility. Nonetheless, knowledge of the direct mediators that synchronously shape the uterine microenvironment for successive yet distinct events, such as the transit of sperm and support for progressive stages of preimplantation embryo development, remain principally deficient. Toward understanding the timed endometrial outputs that permit luminal events as directed by the estrous cycle, we used Bovidae as a model system to uniquely surface sample and study temporal shifts to in vivo endometrial transcripts that encode for proteins destined to be secreted. The results revealed the full quantitative profile of endometrial components that shape the uterine luminal microenvironment at distinct phases of the estrous cycle (estrus, metestrus, diestrus, and proestrus). In interpreting this comprehensive log of stage-specific endometrial secretions, we define the "uterine secretory cycle" and extract a predictive understanding of recurring physiological actions regulated within the uterine lumen in anticipation of sperm and preimplantation embryonic stages. This repetitive microenvironmental preparedness to sequentially provide operative support was a stable intrinsic framework, with only limited responses to sperm or embryos if encountered in the lumen within the cyclic time period. In uncovering the secretory cycle and unraveling realistic biological processes, we present novel foundational knowledge of terminal effectors controlled by the HPG axis to direct a recurring sequence of vital functions within the uterine lumen.NEW & NOTEWORTHY This study unravels the recurring sequence of changes within the uterus that supports vital functions (sperm transit and development of preimplantation embryonic stages) during the reproductive cycle in female Ruminantia. These data present new systems knowledge in uterine reproductive physiology crucial for setting up in vitro biomimicry and artificial environments for assisted reproduction technologies for a range of mammalian species.
Collapse
Affiliation(s)
- Kasey M Schalich
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, New York, United States
| | - Prasanthi P Koganti
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, New York, United States
| | - Juan M Castillo
- Department of Clinical Sciences, Veterinary College, Cornell University, Ithaca, New York, United States
| | - Olivia M Reiff
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, New York, United States
| | - Soon Hon Cheong
- Department of Clinical Sciences, Veterinary College, Cornell University, Ithaca, New York, United States
| | - Vimal Selvaraj
- Department of Animal Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, New York, United States
| |
Collapse
|
48
|
Zolzaya S, Narumoto A, Katsuyama Y. Genomic variation in neurons. Dev Growth Differ 2024; 66:35-42. [PMID: 37855730 DOI: 10.1111/dgd.12898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 10/15/2023] [Accepted: 10/16/2023] [Indexed: 10/20/2023]
Abstract
Neurons born during the fetal period have extreme longevity and survive until the death of the individual because the human brain has highly limited tissue regeneration. The brain is comprised of an enormous variety of neurons each exhibiting different morphological and physiological characteristics and recent studies have further reported variations in their genome including chromosomal abnormalities, copy number variations, and single nucleotide mutations. During the early stages of brain development, the increasing number of neurons generated at high speeds has been proposed to lead to chromosomal instability. Additionally, mutations in the neuronal genome can occur in the mature brain. This observed genomic mosaicism in the brain can be produced by multiple endogenous and environmental factors and careful analyses of these observed variations in the neuronal genome remain central for our understanding of the genetic basis of neurological disorders.
Collapse
Affiliation(s)
- Sunjidmaa Zolzaya
- Division of Neuroanatomy, Department of Anatomy, Shiga University of Medical Science, Otsu, Japan
| | - Ayano Narumoto
- Division of Neuroanatomy, Department of Anatomy, Shiga University of Medical Science, Otsu, Japan
| | - Yu Katsuyama
- Division of Neuroanatomy, Department of Anatomy, Shiga University of Medical Science, Otsu, Japan
| |
Collapse
|
49
|
Moon TS. EBRC: Enhancing bioeconomy through research and communication. N Biotechnol 2023; 78:150-152. [PMID: 37918664 DOI: 10.1016/j.nbt.2023.10.012] [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: 09/12/2023] [Revised: 10/29/2023] [Accepted: 10/30/2023] [Indexed: 11/04/2023]
Abstract
On September 12, 2022, President Biden issued Executive Order 14081 to enable the progress of biomanufacturing and biotechnology. This timely initiative will help overcome many challenging issues, and its potential impacts will be huge. This article discusses eight recommendations to make this US national initiative successful, encourage other nations to consider similar initiatives, and create a better world for the next generations.
Collapse
Affiliation(s)
- Tae Seok Moon
- Moonshot Bio, Inc., 73 Turnpike Street, North Andover, MA 01845, USA.
| |
Collapse
|
50
|
Nagai T. How I overcame problems in in vitro fertilisation of livestock animals. Reprod Fertil Dev 2023; 36:27-33. [PMID: 38064191 DOI: 10.1071/rd23162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023] Open
Abstract
In my research life of 35years, growing with IETS as a researcher of in vitro maturation and fertilisation (IVM/IVF) of porcine and cattle oocytes, I suffered from hard times related to solving problems that prevented the progress of my research and conferment of my degrees. Many researchers may have similar problems. Thus, I would like to address a few examples of how I overcame these problems related to IVF and help young researchers with similar troubles. There were four main problems to be solved in my experiments. Problem 1: Establishment of IVF using only defined medium in pigs. Problem 2: Establishment of successful in vitro culture (IVC) of IVM/IVF bovine oocytes in defined medium. Problem 3: Low rate of male pronucleus formation in IVM porcine oocytes after IVF. Problem 4: Sedimentation of Ca2+ in the sperm capacitation solution for IVF in pigs. Problem 1 was solved by a lucky accident, in which a sperm suspension that would have otherwise been discarded happened to be successfully used for IVF in pigs. Problems 2, 3 and 4 were solved by communication with scientists whose fields were different from mine, where similar problems had been solved already. Young researchers are encouraged to transcend the boundaries of their research fields and solve problems by interacting with researchers in different fields. There are many good connections or answers around us that may be effective in resolving the problems that are hindering the progress of pending research.
Collapse
Affiliation(s)
- Takashi Nagai
- 104 Universe Tsukuba, 2-32-7 Umezono, Tsukuba, Ibaraki 305-0045, Japan
| |
Collapse
|