1
|
Liu X, Ye L, Ding Y, Gong W, Qian H, Jin K, Niu Y, Zuo Q, Song J, Han W, Chen G, Li B. Role of PI3K/AKT signaling pathway involved in self-renewing and maintaining biological properties of chicken primordial germ cells. Poult Sci 2024; 103:104140. [PMID: 39173217 PMCID: PMC11379996 DOI: 10.1016/j.psj.2024.104140] [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: 01/17/2024] [Revised: 06/29/2024] [Accepted: 07/25/2024] [Indexed: 08/24/2024] Open
Abstract
Avian primordial germ cells (PGCs) are important culture cells for the production of transgenic chickens and preservation of the genetic resources of endangered species; however, culturing these cells in vitro proves challenging. Although the proliferation of chicken PGCs is dependent on insulin, the underlying molecular mechanisms remain unclear. In the present study, we explored the expression of the PI3K/AKT signaling pathway in PGCs, investigated its effects on PGC self-renewal and biological properties, and identified the underlying mechanisms. Our findings indicated that although supplementation with the PI3K/AKT activator IGF-1 failed to promote proliferation under the assessed culture conditions, the PI3K/AKT inhibitor LY294002 resulted in retarded cell proliferation and reduced expression of germ cell-related markers. We further demonstrated that inhibition of PI3K/AKT regulates the cell cycle and promotes apoptosis in PGCs by activating the expression of BAX and inhibiting that of Bcl-2. These findings indicated that the PI3K/AKT pathway is required for cell renewal, apoptosis, and maintenance of the reproductive potential in chicken PGCs. This study aimed to provide a theoretical basis for the optimization and improvement of a culture system for chicken PGCs and provide insights into the self-renewal of vertebrate PGCs as well as potential evolutionary changes in this unique cell population.
Collapse
Affiliation(s)
- Xin Liu
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Liu Ye
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Ying Ding
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Wei Gong
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Hongwu Qian
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Kai Jin
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Yingjie Niu
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Qisheng Zuo
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Jiuzhou Song
- Animal & Avian Sciences, University of Maryland, College Park, MA 20742, USA
| | - Wei Han
- Poultry Institute, Chinese Academy of Agricultural Sciences Poultry Institute of Jiangsu, Yangzhou 225003, China
| | - Guohong Chen
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Bichun Li
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China; College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China.
| |
Collapse
|
2
|
Hu T, Purdy PH, Blank MH, Muhonja CK, Pereira RJG, Tiambo CK, McGrew MJ. Direct in vitro propagation of avian germ cells from an embryonic gonad biorepository. Poult Sci 2024; 103:104260. [PMID: 39244782 PMCID: PMC11407075 DOI: 10.1016/j.psj.2024.104260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/05/2024] [Accepted: 08/20/2024] [Indexed: 09/10/2024] Open
Abstract
Direct introduction of cryopreserved embryonic gonadal germ cells (GGC) into a sterile chicken surrogate host to reconstitute a chicken breed has been demonstrated as a feasible approach for preserving and utilizing chicken genetic resources. This method is highly efficient using male gonads; however, a large number of frozen female embryonic gonads is needed to provide sufficient purified GGC for the generation of fertile surrogate female hosts. Applying this method to indigenous chicken breeds and other bird species is difficult due to small flock numbers and poor egg production in each egg laying cycle. Propagating germ cells from the frozen gonadal tissues may be a solution for the biobanking of these birds. Here, we describe a simplified method for culture of GGC from frozen embryonic 9.5 d gonads. At this developmental stage, the germ cells are autonomously shed into medium, yielding hundreds to thousands of mitosis-competent germ cells. The resulting cultures of GGC have over 90% purity, uniformly express SSEA-1 and DAZL antigens and can re-colonize recipient's gonads. The GGC recovery rate from frozen gonads are 42% to 100%, depending on length of cryopreservation and the breed or line of chickens. Entire chicken embryos can also be directly cryopreserved for later gonadal isolation and culture. This storage method is a supplementary approach to safeguard local indigenous chicken breeds bearing valuable genetic traits and should be applicable to the biobanking of many bird species.
Collapse
Affiliation(s)
- Tuanjun Hu
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Edinburgh, EH25 9RG, UK; National Gene Pool of Waterfowl, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou China 225300
| | - Phillip H Purdy
- United States Department of Agriculture (USDA), Agriculture Research Service (ARS), National Animal Germplasm Program, Fort Collins, CO, 80521-4500, USA
| | - Marcel H Blank
- Department of Animal Reproduction, College of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, Sao Paulo, 13635-900, Brazil
| | - Christine K Muhonja
- Centre for Tropical Livestock Genetics and Health (CTLGH), International Livestock Research Institute (ILRI), Box 30709, Nairobi, Kenya; Kenya Agricultural and Livestock Research Organization (KALRO), P. O. Box 25, Naivasha, Kenya
| | - Ricardo J G Pereira
- Department of Animal Reproduction, College of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, Sao Paulo, 13635-900, Brazil
| | - Christian K Tiambo
- Centre for Tropical Livestock Genetics and Health (CTLGH), International Livestock Research Institute (ILRI), Box 30709, Nairobi, Kenya
| | - Mike J McGrew
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Edinburgh, EH25 9RG, UK.
| |
Collapse
|
3
|
Molnár M, Lázár B, Sztán N, Végi B, Drobnyák Á, Buda K, Nagy N, Szőcs E, Pecsenye-Fejszák N, Liptói K, Gócza E, McGrew MJ, Várkonyi E. Successful formation of sperm cells from transplanted primordial germ cells in sterile interspecific avian recipients. Sci Rep 2024; 14:24678. [PMID: 39433810 PMCID: PMC11494066 DOI: 10.1038/s41598-024-74442-0] [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: 05/28/2024] [Accepted: 09/26/2024] [Indexed: 10/23/2024] Open
Abstract
Primordial germ cells (PGCs) are stem cells, from which only gametes develop. In birds, the female sex is heterogametic, thus female gene conservation necessitates preservation of PGCs. PGC transplantation can generate germline chimeras in a host organism and develop into gametes. However, competition between host and transplanted PGCs hinder efficiency of germline chimera generation. We hypothezised that in sterile hybrid recipients with no germ cells of its own, transplanted donor PGCs may exclusively form gametes. Advantages of sterile hybrids as host for PGCs is compliant with many national regulations on genetically modified organisms and technically simpler procedure than the use of busulphan. Therefore, we investigated whether sterile interspecific hybrids may be suitable as recipients for supporting donor PGCs by injecting green fluorescent protein-labelled chicken PGCs into 3-day-old Guinea fowl and domestic fowl hybrid embryos and monitoring PGC development. The injected PGCs colonized almost 100% of the recipient gonads and produced mature spermatozoa after 44 weeks. However, gamete production in these hybrids was initiated much slower than in domestic fowls. This delay may be caused by suboptimal hormonal regulation of gametogenesis in the hybrids. Our results suggest that sterile interspecific hybrids may be suitable hosts for PGCs for efficient gamete production.
Collapse
Affiliation(s)
- Mariann Molnár
- National Centre for Biodiversity and Gene Conservation, Institute for Farm Animal Gene Conservation, Gödöllő, Hungary
| | - Bence Lázár
- National Centre for Biodiversity and Gene Conservation, Institute for Farm Animal Gene Conservation, Gödöllő, Hungary
- Animal Biotechnology Department, Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Gödöllő, Hungary
| | - Nikoletta Sztán
- National Centre for Biodiversity and Gene Conservation, Institute for Farm Animal Gene Conservation, Gödöllő, Hungary
| | - Barbara Végi
- National Centre for Biodiversity and Gene Conservation, Institute for Farm Animal Gene Conservation, Gödöllő, Hungary
| | - Árpád Drobnyák
- National Centre for Biodiversity and Gene Conservation, Institute for Farm Animal Gene Conservation, Gödöllő, Hungary
| | - Kitti Buda
- National Centre for Biodiversity and Gene Conservation, Institute for Farm Animal Gene Conservation, Gödöllő, Hungary
| | - Nándor Nagy
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Emőke Szőcs
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Nóra Pecsenye-Fejszák
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Krisztina Liptói
- National Centre for Biodiversity and Gene Conservation, Institute for Farm Animal Gene Conservation, Gödöllő, Hungary
| | - Elen Gócza
- Animal Biotechnology Department, Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Gödöllő, Hungary
| | - Michael J McGrew
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK
| | - Eszter Várkonyi
- National Centre for Biodiversity and Gene Conservation, Institute for Farm Animal Gene Conservation, Gödöllő, Hungary.
| |
Collapse
|
4
|
Ecker A, Lázár B, Tóth RI, Urbán M, Hoffmann OI, Fekete Z, Barta E, Uher F, Matula Z, Várkonyi E, Gócza E. Creating a novel method for chicken primordial germ cell health monitoring using the fluorescent ubiquitination-based cell cycle indicator reporter system. Poult Sci 2024; 103:104144. [PMID: 39173570 PMCID: PMC11382113 DOI: 10.1016/j.psj.2024.104144] [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: 06/02/2024] [Accepted: 07/25/2024] [Indexed: 08/24/2024] Open
Abstract
The most current in vitro genetic methods, including gene preservation, gene editing and developmental modelling, require a significant number of healthy cells. In poultry species, primordial germ cells (PGCs) are great candidates for all the above-mentioned purposes, given their easy culturing and well-established freezing method for chicken. However, the constant monitoring of cultures can be financially challenging and consumes large amounts of solutions and accessories. This study aimed to introduce the Fluorescent Ubiquitination-based Cell Cycle Indicator (FUCCI) complex into the chicken PGCs. FUCCI is a powerful transgenic tool based on the periodic protein expression changes during the cell cycle. It includes chromatin licensing and DNA replication factor 1 attached monomeric Kusabira-Orange and Geminin-attached monomeric Azami-Green fluorescent proteins, that cause the cells to express a red signal in the G1 phase and a green signal in S and G2 phases. Modification of the chicken PGCs was done via electroporation and deemed to be successful according to confocal microscopy, DNA sequencing and timelapse video analysis. Stable clone cell lines were established, cryopreserved, and injected into recipient embryos to prove the integrational competency. The cell health monitoring was tested with medium change experiments, that proved the intended reactions of the FUCCI transgene. These results established the future for FUCCI experiments in chicken, including heat treatment and toxin treatment.
Collapse
Affiliation(s)
- András Ecker
- Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Gödöllő, 2100 Hungary; Agribiotechnology and Precision Breeding for Food Security National Laboratory, Gödöllő, 2100 Hungary
| | - Bence Lázár
- Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Gödöllő, 2100 Hungary; Agribiotechnology and Precision Breeding for Food Security National Laboratory, Gödöllő, 2100 Hungary; National Centre for Biodiversity and Gene Conservation, Gödöllő, 2100 Hungary
| | - Roland I Tóth
- Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Gödöllő, 2100 Hungary; Agribiotechnology and Precision Breeding for Food Security National Laboratory, Gödöllő, 2100 Hungary
| | - Martin Urbán
- Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Gödöllő, 2100 Hungary; Agribiotechnology and Precision Breeding for Food Security National Laboratory, Gödöllő, 2100 Hungary
| | - Orsolya I Hoffmann
- Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Gödöllő, 2100 Hungary; Agribiotechnology and Precision Breeding for Food Security National Laboratory, Gödöllő, 2100 Hungary
| | - Zsófia Fekete
- Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Gödöllő, 2100 Hungary; Department of Environmental and Biological Sciences, University of Eastern Finland, Joensuu, 80101 Finland
| | - Endre Barta
- Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Gödöllő, 2100 Hungary; Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, 4032 Hungary
| | - Ferenc Uher
- National Institute of Hematology and Infectology, Budapest, 1097 Hungary
| | - Zsolt Matula
- National Institute of Hematology and Infectology, Budapest, 1097 Hungary
| | - Eszter Várkonyi
- National Centre for Biodiversity and Gene Conservation, Gödöllő, 2100 Hungary
| | - Elen Gócza
- Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Gödöllő, 2100 Hungary; Agribiotechnology and Precision Breeding for Food Security National Laboratory, Gödöllő, 2100 Hungary.
| |
Collapse
|
5
|
Kinoshita K, Tanabe K, Nakamura Y, Nishijima KI, Suzuki T, Okuzaki Y, Mizushima S, Wang MS, Khan SU, Xu K, Jamal MA, Wei T, Zhao H, Su Y, Sun F, Liu G, Zhu F, Zhao HY, Wei HJ. PGC-based cryobanking, regeneration through germline chimera mating, and CRISPR/Cas9-mediated TYRP1 modification in indigenous Chinese chickens. Commun Biol 2024; 7:1127. [PMID: 39271811 PMCID: PMC11399235 DOI: 10.1038/s42003-024-06775-5] [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: 03/01/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
Primordial germ cells (PGCs) are vital for producing sperm and eggs and are crucial for conserving chicken germplasm and creating genetically modified chickens. However, efforts to use PGCs for preserving native chicken germplasm and genetic modification via CRISPR/Cas9 are limited. Here we show that we established 289 PGC lines from eight Chinese chicken populations with an 81.6% success rate. We regenerated Piao chickens by repropagating cryopreserved PGCs and transplanting them into recipient chickens, achieving a 12.7% efficiency rate. These regenerated chickens carried mitochondrial DNA from female donor PGC and the rumplessness mutation from both male and female donors. Additionally, we created the TYRP1 (tyrosinase-related protein 1) knockout (KO) PGC lines via CRISPR/Cas9. Transplanting KO cells into male recipients and mating them with wild-type hens produced four TYRP1 KO chickens with brown plumage due to reduced eumelanin production. Our work demonstrates efficient PGC culture, cryopreservation, regeneration, and gene editing in chickens.
Collapse
Affiliation(s)
- Keiji Kinoshita
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, Yunnan, 650201, China
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, 650201, China
| | - Kumiko Tanabe
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, Yunnan, 650201, China
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, 650201, China
| | - Yoshiaki Nakamura
- Laboratory of Animal Breeding and Genetics, Graduate School of Integrated Sciences for Life and School of Applied Biological Science, Hiroshima University, Hiroshima, 739-8528, Japan
| | - Ken-Ichi Nishijima
- Avian Bioscience Research Center, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, 464-8601, Japan
| | - Takayuki Suzuki
- Department of Biology, Graduate School of Science, Osaka Metropolitan University, Osaka, 558-8585, Japan
| | - Yuya Okuzaki
- Avian Bioscience Research Center, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, 464-8601, Japan
| | - Shusei Mizushima
- Department of Biological Sciences, Faculty of Science, Hokkaido University, Sapporo, 060-0810, Japan
| | - Ming-Shan Wang
- State Key Laboratory of Genetic resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Sami Ullah Khan
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, Yunnan, 650201, China
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, 650201, China
| | - Kaixiang Xu
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, Yunnan, 650201, China
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, 650201, China
| | - Muhammad Ameen Jamal
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, Yunnan, 650201, China
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, 650201, China
- State Key Laboratory of Genetic resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Taiyun Wei
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, Yunnan, 650201, China
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, 650201, China
| | - Heng Zhao
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, Yunnan, 650201, China
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, 650201, China
| | - Yanhua Su
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, Yunnan, 650201, China
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, 650201, China
| | - Feizhou Sun
- National Center for Preservation of Animal Genetic Resources, National Animal Husbandry Service, Beijing, 100125, China
| | - Gang Liu
- National Center for Preservation of Animal Genetic Resources, National Animal Husbandry Service, Beijing, 100125, China
| | - Fangxian Zhu
- National Center for Preservation of Animal Genetic Resources, National Animal Husbandry Service, Beijing, 100125, China
| | - Hong-Ye Zhao
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, Yunnan, 650201, China.
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, 650201, China.
| | - Hong-Jiang Wei
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, Yunnan, 650201, China.
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, 650201, China.
- State Key Laboratory of Genetic resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.
| |
Collapse
|
6
|
Oh JDH, Freem L, Saunders DDZ, McTeir L, Gilhooley H, Jackson M, Glover JD, Smith J, Schoenebeck JJ, Lettice LA, Sang HM, Davey MG. Insights into digit evolution from a fate map study of the forearm using Chameleon, a new transgenic chicken line. Development 2024; 151:dev202340. [PMID: 38828852 PMCID: PMC11234372 DOI: 10.1242/dev.202340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 05/20/2024] [Indexed: 06/05/2024]
Abstract
The cellular and genetic networks that contribute to the development of the zeugopod (radius and ulna of the forearm, tibia and fibula of the leg) are not well understood, although these bones are susceptible to loss in congenital human syndromes and to the action of teratogens such as thalidomide. Using a new fate-mapping approach with the Chameleon transgenic chicken line, we show that there is a small contribution of SHH-expressing cells to the posterior ulna, posterior carpals and digit 3. We establish that although the majority of the ulna develops in response to paracrine SHH signalling in both the chicken and mouse, there are differences in the contribution of SHH-expressing cells between mouse and chicken as well as between the chicken ulna and fibula. This is evidence that, although zeugopod bones are clearly homologous according to the fossil record, the gene regulatory networks that contribute to their development and evolution are not fixed.
Collapse
Affiliation(s)
- Julia Dong Hwa Oh
- Functional Genetics, The Roslin Institute R(D)SVS, CMVM, University of Edinburgh, Edinburgh EH25 9RG, UK
| | - Lu Freem
- Functional Genetics, The Roslin Institute R(D)SVS, CMVM, University of Edinburgh, Edinburgh EH25 9RG, UK
| | - Dillan D. Z. Saunders
- Functional Genetics, The Roslin Institute R(D)SVS, CMVM, University of Edinburgh, Edinburgh EH25 9RG, UK
| | - Lynn McTeir
- Functional Genetics, The Roslin Institute R(D)SVS, CMVM, University of Edinburgh, Edinburgh EH25 9RG, UK
| | - Hazel Gilhooley
- Functional Genetics, The Roslin Institute R(D)SVS, CMVM, University of Edinburgh, Edinburgh EH25 9RG, UK
| | - Melany Jackson
- Genetics and Genomics, The Roslin Institute R(D)SVS, CMVM, University of Edinburgh, Edinburgh EH25 9RG, UK
| | - James D. Glover
- Functional Genetics, The Roslin Institute R(D)SVS, CMVM, University of Edinburgh, Edinburgh EH25 9RG, UK
| | - Jonathan Smith
- Functional Genetics, The Roslin Institute R(D)SVS, CMVM, University of Edinburgh, Edinburgh EH25 9RG, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Jeffrey J. Schoenebeck
- Genetics and Genomics, The Roslin Institute R(D)SVS, CMVM, University of Edinburgh, Edinburgh EH25 9RG, UK
| | - Laura A. Lettice
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Helen M. Sang
- Functional Genetics, The Roslin Institute R(D)SVS, CMVM, University of Edinburgh, Edinburgh EH25 9RG, UK
| | - Megan G. Davey
- Functional Genetics, The Roslin Institute R(D)SVS, CMVM, University of Edinburgh, Edinburgh EH25 9RG, UK
| |
Collapse
|
7
|
Ibrahim M, Grochowska E, Lázár B, Várkonyi E, Bednarczyk M, Stadnicka K. The Effect of Short- and Long-Term Cryopreservation on Chicken Primordial Germ Cells. Genes (Basel) 2024; 15:624. [PMID: 38790253 PMCID: PMC11121574 DOI: 10.3390/genes15050624] [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: 05/10/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
Primordial germ cells (PGCs) are the precursors of functional gametes and the only cell type capable of transmitting genetic and epigenetic information from generation to generation. These cells offer valuable starting material for cell-based genetic engineering and genetic preservation, as well as epigenetic studies. While chicken PGCs have demonstrated resilience in maintaining their germness characteristics during both culturing and cryopreservation, their handling remains a complex challenge requiring further refinement. Herein, the study aimed to compare the effects of different conditions (freezing-thawing and in vitro cultivation) on the expression of PGC-specific marker genes. Embryonic blood containing circulating PGCs was isolated from purebred Green-legged Partridgelike chicken embryos at 14-16 Hamburger-Hamilton (HH) embryonic development stage. The blood was pooled separately for males and females following sex determination. The conditions applied to the blood containing PGCs were as follows: (1) fresh isolation; (2) cryopreservation for a short term (2 days); and (3) in vitro culture (3 months) with long-term cryopreservation of purified PGCs (~2 years). To characterize PGCs, RNA isolation was carried out, followed by quantitative reverse transcription polymerase chain reaction (RT-qPCR) to assess the expression levels of specific germ cell markers (SSEA1, CVH, and DAZL), as well as pluripotency markers (OCT4 and NANOG). The investigated genes exhibited consistent expression among PGCs maintained under diverse conditions, with no discernible differences observed between males and females. Notably, the analyzed markers demonstrated higher expression levels in PGCs when subjected to freezing than in their freshly isolated counterparts.
Collapse
Affiliation(s)
- Mariam Ibrahim
- Department of Animal Biotechnology and Genetics, Bydgoszcz University of Science and Technology, Mazowiecka 28, 85-084 Bydgoszcz, Poland
- PBS Doctoral School, Bydgoszcz University of Science and Technology, Aleje Prof. S. Kaliskiego 7, 85-796 Bydgoszcz, Poland
| | - Ewa Grochowska
- Department of Animal Biotechnology and Genetics, Bydgoszcz University of Science and Technology, Mazowiecka 28, 85-084 Bydgoszcz, Poland
| | - Bence Lázár
- National Centre for Biodiversity and Gene Conservation, Institute for Farm Animal Gene Conservation, Isaszegi Street 200, 2100 Godollo, Hungary
- Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Szent-Gyorgyi Albert Street 4, 2100 Godollo, Hungary
| | - Eszter Várkonyi
- National Centre for Biodiversity and Gene Conservation, Institute for Farm Animal Gene Conservation, Isaszegi Street 200, 2100 Godollo, Hungary
| | - Marek Bednarczyk
- Department of Animal Biotechnology and Genetics, Bydgoszcz University of Science and Technology, Mazowiecka 28, 85-084 Bydgoszcz, Poland
| | - Katarzyna Stadnicka
- Faculty of Health Sciences, Collegium Medicum, Nicolaus Copernicus University, Łukasiewicza 1, 85-821 Bydgoszcz, Poland
| |
Collapse
|
8
|
Ichikawa K, McGrew MJ. Innovations in poultry reproduction using cryopreserved avian germ cells. Reprod Domest Anim 2024; 59:e14591. [PMID: 38798199 DOI: 10.1111/rda.14591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/19/2024] [Accepted: 05/03/2024] [Indexed: 05/29/2024]
Abstract
Meat and eggs from chicken are the major source of animal protein for the human population. The cryopreservation of poultry species is needed to guarantee sustainable production. Here, we describe the existing cryopreservation technologies for avian reproductive cells using embryonic germ cells, spermatozoa and ovarian tissues. We outline strategies to reconstitute chicken breeds from their cryopreserved embryonic germ cells using surrogate hosts and discuss the perspectives for genetic conservation and reconstitution of chicken and wild avian species using surrogate host animals.
Collapse
Affiliation(s)
- Kennosuke Ichikawa
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Mike J McGrew
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| |
Collapse
|
9
|
Blank MH, Kawaoku AJT, Rui BR, Carreira ACO, Hamilton TRDS, Goissis MD, Pereira RJG. Successful xenotransplantation of testicular cells following fractionated chemotherapy of recipient birds. Sci Rep 2024; 14:3085. [PMID: 38321093 PMCID: PMC10847125 DOI: 10.1038/s41598-023-45019-0] [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: 05/19/2023] [Accepted: 10/14/2023] [Indexed: 02/08/2024] Open
Abstract
An essential step in the success of germ cell transplantation is the preparation of the recipient's testicular environment to increase the availability of stem cell niches. However, most methods for this purpose in birds face serious limitations such as partial germ cell depletion, high toxicity and mortality, or the need to use expensive technologies. Here, we validated a simple and practical technique of transferring quail testicular cells into chicken testes depleted of endogenous spermatozoa by fractioned chemotherapy (20 mg/kg/week busulfan for 5 weeks). This protocol resulted in a very low mortality of the treated day-old chicks and, despite maintenance of androgenic activity, sperm production was decreased by 84.3% at 25 weeks of age. NANOG immunostaining revealed that very few to no germ cells were present following treatment with 20 and 40 mg/kg, respectively. RT-qPCR data also showed that c-MYC and NANOG expression declined in these treatments, but GRFα1 and BID expressions remained unaltered among groups. After xenotransplantation, quail germ cells were immunodetected in chicken testes using a species-specific antibody (QCPN), and quail ovalbumin DNA was found in seminal samples collected from chicken recipients. Together, these data confirm that fractionated administration of busulfan in hatchlings is a practical, effective, and safe protocol to prepare recipient male birds capable of supporting xenogeneic spermatogenesis.
Collapse
Affiliation(s)
- Marcel Henrique Blank
- Department of Animal Reproduction, College of Veterinary Medicine and Animal Science, University of São Paulo, Av. Duque de Caxias Norte 255, Pirassununga, SP , CEP 13635-900, Brazil.
| | | | - Bruno Rogério Rui
- Department of Animal Reproduction, College of Veterinary Medicine and Animal Science, University of São Paulo, Av. Duque de Caxias Norte 255, Pirassununga, SP , CEP 13635-900, Brazil
| | - Ana Claudia Oliveira Carreira
- Cell and Molecular Therapy Center (NUCEL), Medical School, University of Sao Paulo, Rua Pangaré 100, São Paulo, 05360-130, Brazil
| | - Thais Rose Dos Santos Hamilton
- Department of Animal Reproduction, College of Veterinary Medicine and Animal Science, University of São Paulo, Av. Duque de Caxias Norte 255, Pirassununga, SP , CEP 13635-900, Brazil
| | - Marcelo Demarchi Goissis
- Department of Animal Reproduction, College of Veterinary Medicine and Animal Science, University of São Paulo, Av. Duque de Caxias Norte 255, Pirassununga, SP , CEP 13635-900, Brazil
| | - Ricardo José Garcia Pereira
- Department of Animal Reproduction, College of Veterinary Medicine and Animal Science, University of São Paulo, Av. Duque de Caxias Norte 255, Pirassununga, SP , CEP 13635-900, Brazil.
| |
Collapse
|
10
|
Chojnacka-Puchta L, Sawicka D. Purification and Transfection Methods of Chicken Primordial Germ Cells. Methods Mol Biol 2024; 2770:15-25. [PMID: 38351443 DOI: 10.1007/978-1-0716-3698-5_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2024]
Abstract
Primordial germ cells (PGCs) play a special role in the vertebrate life cycle since they are the precursors of germ cells through which the genome is passed to the next generations. PGCs are found in different locations and variable numbers in the chick embryo, as in other species, depending on the developmental stages. Here, we describe in detail a method based on the Percoll gradient, routinely used in our laboratory, allowing us to obtain from blood and gonad anlages significant numbers of viable PGCs which can be successfully cultured or efficiently genetically modified.
Collapse
Affiliation(s)
- Luiza Chojnacka-Puchta
- Department of Chemical, Biological and Aerosol Hazards, Central Institute for Labour Protection-National Research Institute, Warsaw, Poland.
| | - Dorota Sawicka
- Department of Chemical, Biological and Aerosol Hazards, Central Institute for Labour Protection-National Research Institute, Warsaw, Poland.
| |
Collapse
|
11
|
Sritabtim K, Prukudom S, Piyasanti Y, Chaipipat S, Kuwana T, Jurutha J, Sinsiri R, Tirawattanawanich C, Siripattarapravat K. First study on repeatable culture of primordial germ cells from various embryonic regions with giant feeder cells in Japanese quail (Coturnix japonica). Theriogenology 2024; 213:43-51. [PMID: 37797528 DOI: 10.1016/j.theriogenology.2023.09.020] [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: 08/04/2023] [Revised: 09/08/2023] [Accepted: 09/24/2023] [Indexed: 10/07/2023]
Abstract
Japanese quail (JQ, Coturnix japonica) is a farmed animal with a high economic value and has been used extensively as an avian model for research. Germline chimera production based on cryopreserved primordial germ cells (PGCs) is possible for conservation management of quail breeds as successful isolation has been reported of PGCs from their blood and gonads. However, the repeatable cultivation protocol has not been elucidated yet, which has hindered technological development. The current study characterized cultivation of pregonadal PGCs isolated from embryonic parts; embryonic blood (cPGCs), whole embryonic tissues (tPGCs), parts of tail buds (tbPGCs), and a mixture of blood and tail bud tissues (ctbPGCs). The results showed that the cultivation system required the presence of specific embryonic cells to act as a feeder for JQ-PGCs and that such a system facilitated more successful cultivation, as shown by the percentages of isolation and cultivation in tbPGCs (100%, 100%, respectively), tPGCs (60%, 55%, respectively), and ctbPGCs (60%, 30%, respectively), but not in cPGCs (0%) cultured on a mitomycin-treated JQ feeder cell-line. Once the co-culture system had been established, the PGCs could be propagated for at least 5 months. These PGCs expressed germ cell-specific markers (DAZL and CVH) and could colonize embryonic gonads. Conclusively, the isolation of pregonadal PGCs and their long-term cultivation in vitro requires a unique embryonic cell, giant cell feeder, that is indispensable for the proliferation of PGCs. Characterization of cell signaling sustaining a mutual interaction between the PGCs and the specific feeder cells will elucidate a superior environment for in vitro cultivation, as well as support the minimal transfer of used xenobiotics in chimera production.
Collapse
Affiliation(s)
- Kornkanok Sritabtim
- Center for Veterinary Diagnostic Laboratory - Bangkhen, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, Thailand
| | - Sukumal Prukudom
- Center for Veterinary Diagnostic Laboratory - Bangkhen, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, Thailand; Department of Anatomy, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, Thailand
| | - Yanika Piyasanti
- Center for Veterinary Diagnostic Laboratory - Bangkhen, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, Thailand
| | - Suparat Chaipipat
- Center for Veterinary Diagnostic Laboratory - Bangkhen, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, Thailand; Center for Agricultural Biotechnology, Kasetsart University, Kamphaengsaen Campus, Nakhon Pathom, Thailand; Center of Excellence on Agricultural Biotechnology:(AG-BIO/PERDO-CHE), Bangkok, Thailand
| | | | - Juthathip Jurutha
- Center for Veterinary Diagnostic Laboratory - Bangkhen, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, Thailand
| | - Rungthiwa Sinsiri
- Center for Veterinary Diagnostic Laboratory - Bangkhen, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, Thailand
| | - Chanin Tirawattanawanich
- Center for Veterinary Diagnostic Laboratory - Bangkhen, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, Thailand
| | - Kannika Siripattarapravat
- Center for Veterinary Diagnostic Laboratory - Bangkhen, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, Thailand; Center for Agricultural Biotechnology, Kasetsart University, Kamphaengsaen Campus, Nakhon Pathom, Thailand; Center of Excellence on Agricultural Biotechnology:(AG-BIO/PERDO-CHE), Bangkok, Thailand; Department of Pathology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, Thailand.
| |
Collapse
|
12
|
Zhang X, Xian R, Fu Y, Dai Y, Peng R. A Novel, Efficient Method to Isolate Chicken Primordial Germ Cells from Embryonic Blood Using Cell Culture Inserts. Animals (Basel) 2023; 13:3805. [PMID: 38136842 PMCID: PMC10740788 DOI: 10.3390/ani13243805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/01/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
Primordial germ cells (PGCs) play a crucial role in preserving poultry genetic resources and conducting transgenic research. A system for the rapid isolation of PGCs from single chicken embryonic blood was established in this paper. We found that PGCs can migrate to the lower layer of chicken embryonic fibroblasts (CEFs) through pores smaller than their diameter, while blood cells cannot, when co-cultured with CEFs of passages two to three. Based on the characteristics of PGCs, we developed a new PGC isolation method (cell culture insert/CEF adhesion method) that utilizes a 3 μm cell culture insert and CEFs of passages two to three. Using this method, approximately 700 PGCs can be isolated from the blood of a single chicken embryo at Hamburger and Hamilton (H&H) stage 17 of development. The separation rate achieved was 87.5%, with a separation purity of 95%. The separation rate of this method was 41.4% higher than the common Percoll density gradient centrifugation method and 33.6% higher than lysis with ACK buffer. PGCs isolated from embryonic blood could proliferate 37-fold within 2 weeks when cultured in a feeder-free culture system. They also continued to express the SSEA-1 and DAZL proteins and retained the ability to migrate in vivo. Overall, PGCs separated using cell culture inserts/CEF adhesion method retain their stem cell characteristics and migration ability. PGCs also exhibit good proliferation efficiency, making them suitable for subsequent transgenic experiments or genetic resource preservation.
Collapse
Affiliation(s)
| | | | | | | | - Rui Peng
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610064, China
| |
Collapse
|
13
|
Watanabe T, Ochi Y, Kajihara R, Ichikawa K, Ezaki R, Matsuzaki M, Horiuchi H. Lipofection with Lipofectamine™ 2000 in a heparin-free growth medium results in high transfection efficiency in chicken primordial germ cells. Biotechnol J 2023; 18:e2300328. [PMID: 37559489 DOI: 10.1002/biot.202300328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/31/2023] [Accepted: 08/04/2023] [Indexed: 08/11/2023]
Abstract
Primordial germ cells (PGCs) that can differentiate into gametes are used to produce genome-edited chickens. However, the transfection efficiency into PGCs is low in chickens; therefore, the yield efficiency of PGCs modified via genome editing is problematic. In this study, we improved transfection efficiency and achieved highly efficient genome editing in chicken PGCs. For transfection, we used lipofection, which is convenient for gene transfer. Chicken PGC cultures require adding heparin to support growth; however, heparin significantly reduces lipofection efficiency (p < 0.01). Heparin-induced lipofection efficiency was restored by adding protamine. Based on these results, we optimized gene transfer into chicken PGCs. Lipofectamine 2000 and our PGC medium were the most efficient transfection reagent and medium, respectively. Finally, based on established conditions, we compared the gene knock-out efficiencies of ovomucoid, a major egg allergen, and gene knock-in efficiencies at the ACTB locus. These results indicate that optimized lipofection is useful for CRISPR/Cas9-mediated knock-out and knock-in. Our findings may contribute to the generation of genome-edited chickens and stimulate research in various applications involving them.
Collapse
Affiliation(s)
- Tenkai Watanabe
- Laboratory of Immunobiology, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - Yuta Ochi
- Laboratory of Immunobiology, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - Ryota Kajihara
- Laboratory of Immunobiology, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - Kennosuke Ichikawa
- Genome Editing Innovation Center, Hiroshima University, Higashi-Hiroshima, Japan
- The Roslin Institute, University of Edinburgh, Edinburgh, UK
| | - Ryo Ezaki
- Laboratory of Immunobiology, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - Mei Matsuzaki
- Laboratory of Immunobiology, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - Hiroyuki Horiuchi
- Laboratory of Immunobiology, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
- Genome Editing Innovation Center, Hiroshima University, Higashi-Hiroshima, Japan
| |
Collapse
|
14
|
Intarapat S, Sukparangsi W, Gusev O, Sheng G. A Bird's-Eye View of Endangered Species Conservation: Avian Genomics and Stem Cell Approaches for Green Peafowl ( Pavo muticus). Genes (Basel) 2023; 14:2040. [PMID: 38002983 PMCID: PMC10671381 DOI: 10.3390/genes14112040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
Aves ranks among the top two classes for the highest number of endangered and extinct species in the kingdom Animalia. Notably, the IUCN Red List classified the green peafowl as endangered. This highlights promising strategies using genetics and reproductive technologies for avian wildlife conservation. These platforms provide the capacity to predict population trends and enable the practical breeding of such species. The conservation of endangered avian species is facilitated through the application of genomic data storage and analysis. Storing the sequence is a form of biobanking. An analysis of sequence can identify genetically distinct individuals for breeding. Here, we reviewed avian genomics and stem cell approaches which not only offer hope for saving endangered species, such as the green peafowl but also for other birds threatened with extinction.
Collapse
Affiliation(s)
- Sittipon Intarapat
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Woranop Sukparangsi
- Department of Biology, Faculty of Science, Burapha University, Chonburi 20131, Thailand;
| | - Oleg Gusev
- Regulatory Genomics Research Center, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia;
- Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
- Life Improvement by Future Technologies (LIFT) Center, 143025 Moscow, Russia
| | - Guojun Sheng
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan;
| |
Collapse
|
15
|
Challagulla A, Jenkins KA, O'Neil TE, Morris KR, Wise TG, Tizard ML, Bean AGD, Schat KA, Doran TJ. Germline engineering of the chicken genome using CRISPR/Cas9 by in vivo transfection of PGCs. Anim Biotechnol 2023; 34:775-784. [PMID: 32707002 DOI: 10.1080/10495398.2020.1789869] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Development of simple and readily adoptable methods to mediate germline engineering of the chicken genome will have many applications in research, agriculture and industrial biotechnology. We report germline targeting of the endogenous chicken Interferon Alpha and Beta Receptor Subunit 1 (IFNAR1) gene by in vivo transgenic expression of the high-fidelity Cas9 (Cas9-HF1) and guide RNAs (gRNAs) in chickens. First, we developed a Tol2 transposon vector carrying Cas9-HF1, IFNAR1-gRNAs (IF-gRNAs) and green fluorescent protein (GFP) transgenes (pTgRCG) and validated in chicken fibroblast DF1 cells. Next, the pTgRCG plasmid was directly injected into the dorsal aorta of embryonic day (ED) 2.5 chicken embryos targeting the circulating primordial germ cells (PGCs). The resulting chimera roosters generated a fully transgenic generation 1 (G1) hen with constitutive expression of Cas9-HF1 and IF-gRNAs (G1_Tol2-Cas9/IF-gRNA). We detected a spectrum of indels at gRNA-targeted loci in the G1_Tol2-Cas9/IF-gRNA hen and the indels were stably inherited by the G2 progeny. Breeding of the G1_Tol2-Cas9/IF-gRNA hen resulted in up to 10% transgene-free heterozygote IFNAR1 mutants, following null-segregation of the Tol2 insert. The method described here will provide new opportunities for genome editing in chicken and other avian species that lack PGC culture.
Collapse
Affiliation(s)
- Arjun Challagulla
- Australian Centre for Disease Preparedness, CSIRO Health and Biosecurity, Geelong, Australia
| | - Kristie A Jenkins
- Australian Centre for Disease Preparedness, CSIRO Health and Biosecurity, Geelong, Australia
| | - Terri E O'Neil
- Australian Centre for Disease Preparedness, CSIRO Health and Biosecurity, Geelong, Australia
| | - Kirsten R Morris
- Australian Centre for Disease Preparedness, CSIRO Health and Biosecurity, Geelong, Australia
| | - Terry G Wise
- Australian Centre for Disease Preparedness, CSIRO Health and Biosecurity, Geelong, Australia
| | - Mark L Tizard
- Australian Centre for Disease Preparedness, CSIRO Health and Biosecurity, Geelong, Australia
| | - Andrew G D Bean
- Australian Centre for Disease Preparedness, CSIRO Health and Biosecurity, Geelong, Australia
| | - Karel A Schat
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Timothy J Doran
- Australian Centre for Disease Preparedness, CSIRO Health and Biosecurity, Geelong, Australia
| |
Collapse
|
16
|
Blackburn HD, Azevedo HC, Purdy PH. Incorporation of Biotechnologies into Gene Banking Strategies to Facilitate Rapid Reconstitution of Populations. Animals (Basel) 2023; 13:3169. [PMID: 37893893 PMCID: PMC10603745 DOI: 10.3390/ani13203169] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/05/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
National animal gene banks that are responsible for conserving livestock, poultry, and aquatic genetic resources need to be capable of utilizing a broad array of cryotechnologies coupled with assisted reproductive technologies to reconstitute either specific animals or populations/breeds as needed. This capability is predicated upon having sufficient genetic diversity (usually encapsulated by number of animals in the collection), units of germplasm or tissues, and the ability to reconstitute animals. While the Food and Agriculture Organization of the United Nations (FAO 2012, 2023) developed a set of guidelines for gene banks on these matters, those guidelines do not consider applications and utilization of newer technologies (e.g., primordial germ cells, cloning from somatic cells, embryo transfer, IVF, sex-sorted semen), which can radically change how gene banks collect, store, and utilize genetic resources. This paper reviews the current status of using newer technologies, explores how gene banks might make such technologies part of their routine operations, and illustrates how combining newer assisted reproductive technologies with older approaches enables populations to be reconstituted more efficiently.
Collapse
Affiliation(s)
- Harvey D. Blackburn
- USDA ARS National Animal Germplasm Program, 1111 S. Mason St., Fort Collins, CO 80521-4500, USA
| | | | - Phillip H. Purdy
- USDA ARS National Animal Germplasm Program, 1111 S. Mason St., Fort Collins, CO 80521-4500, USA
| |
Collapse
|
17
|
Golkar-Narenji A, Dziegiel P, Kempisty B, Petitte J, Mozdziak PE, Bryja A. In vitro culture of reptile PGCS to preserve endangered species. Cell Biol Int 2023; 47:1314-1326. [PMID: 37178380 DOI: 10.1002/cbin.12033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 04/05/2023] [Accepted: 05/01/2023] [Indexed: 05/15/2023]
Abstract
Primordial germ cells (PGCs), are the source of gametes in vertebrates. There are similarities in the development of PGCs of reptiles with avian and mammalian species PGCs development. PGCs culture has been performed for avian and mammalian species but there is no report for reptilian PGCs culture. In vitro culture of PGCs is needed to produce transgenic animals, preservation of endangered animals and for studies on cell behaviour and research on fertility. Reptiles are traded as exotic pets and a source of food and they are valuable for their skin and they are useful as model for medical research. Transgenic reptile has been suggested to be useful for pet industry and medical research. In this research different aspects of PGCs development was compared in three main classes of vertebrates including mammalian, avian and reptilian species. It is proposed that a discussion on similarities between reptilian PGCs development with avian and mammalian species helps to find clues for studies of reptilian PGCs development details and finding an efficient protocol for in vitro culture of reptilian PG.
Collapse
Affiliation(s)
- Afsaneh Golkar-Narenji
- Prestage Department of Poultry Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Piotr Dziegiel
- Department of Human Morphology and Embryology, Division of Histology and Embryology, Wrocław Medical University, Wroclaw, Dolnoslaskie, Poland
| | - Bartosz Kempisty
- Prestage Department of Poultry Sciences, North Carolina State University, Raleigh, North Carolina, USA
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Toruń, Poland
- Graduate Physiology Program NC State University North Carolina State University, Raleigh, North Carolina, USA
- Department of Human Morphology and Embryology, Division of Anatomy, Wroclaw Medical University, Wroclaw, Dolnoslaskie, Poland
| | - James Petitte
- Prestage Department of Poultry Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Paul Edward Mozdziak
- Prestage Department of Poultry Sciences, North Carolina State University, Raleigh, North Carolina, USA
- Graduate Physiology Program NC State University North Carolina State University, Raleigh, North Carolina, USA
| | - Artur Bryja
- Department of Human Morphology and Embryology, Division of Anatomy, Wroclaw Medical University, Wroclaw, Dolnoslaskie, Poland
| |
Collapse
|
18
|
Zare M, Mirhoseini SZ, Ghovvati S, Yakhkeshi S, Hesaraki M, Barati M, Sayyahpour FA, Baharvand H, Hassani SN. The constitutively active pSMAD2/3 relatively improves the proliferation of chicken primordial germ cells. Mol Reprod Dev 2023. [PMID: 37379342 DOI: 10.1002/mrd.23689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 05/06/2023] [Accepted: 05/15/2023] [Indexed: 06/30/2023]
Abstract
In many multicellular organisms, mature gametes originate from primordial germ cells (PGCs). Improvements in the culture of PGCs are important not only for developmental biology research, but also for preserving endangered species, and for genome editing and transgenic animal technologies. SMAD2/3 appear to be powerful regulators of gene expression; however, their potential positive impact on the regulation of PGC proliferation has not been taken into consideration. Here, the effect of TGF-β signaling as the upstream activator of SMAD2/3 transcription factors was evaluated on chicken PGCs' proliferation. For this, chicken PGCs at stages 26-28 Hamburger-Hamilton were obtained from the embryonic gonadal regions and cultured on different feeders or feeder-free substrates. The results showed that TGF-β signaling agonists (IDE1 and Activin-A) improved PGC proliferation to some extent while treatment with SB431542, the antagonist of TGF-β, disrupted PGCs' proliferation. However, the transfection of PGCs with constitutively active SMAD2/3 (SMAD2/3CA) resulted in improved PGC proliferation for more than 5 weeks. The results confirmed the interactions between overexpressed SMAD2/3CA and pluripotency-associated genes NANOG, OCT4, and SOX2. According to the results, the application of SMAD2/3CA could represent a step toward achieving an efficient expansion of avian PGCs.
Collapse
Affiliation(s)
- Masumeh Zare
- Department of Animal Sciences, Faculty of Agriculture, University of Guilan, Rasht, Guilan, Iran
| | | | - Shahrokh Ghovvati
- Department of Animal Sciences, Faculty of Agriculture, University of Guilan, Rasht, Guilan, Iran
| | - Saeed Yakhkeshi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahdi Hesaraki
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mojgan Barati
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Forough Azam Sayyahpour
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Seyedeh-Nafiseh Hassani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
19
|
Doddamani D, Woodcock M, Taylor L, Nandi S, McTeir L, Davey MG, Smith J, McGrew MJ. The Transcriptome of Chicken Migratory Primordial Germ Cells Reveals Intrinsic Sex Differences and Expression of Hallmark Germ Cell Genes. Cells 2023; 12:cells12081151. [PMID: 37190060 DOI: 10.3390/cells12081151] [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: 02/26/2023] [Revised: 04/03/2023] [Accepted: 04/09/2023] [Indexed: 05/17/2023] Open
Abstract
Primordial germ cells (PGCs) are germline-restricted embryonic cells that form the functional gametes of the adult animal. The use of avian PGCs in biobanking and producing genetically modified birds has driven research on the in vitro propagation and manipulation of these embryonic cells. In avian species, PGCs are hypothesized to be sexually undetermined at an early embryonic stage and undergo differentiation into an oocyte or spermatogonial fate dictated by extrinsic factors present in the gonad. However, chicken male and female PGCs require different culture conditions, suggesting that there are sex-specific differences, even at early stages. To understand potential differences between male and female chicken PGCs during migratory stages, we studied the transcriptomes of circulatory stage male and female PGCs propagated in a serum-free medium. We found that in vitro cultured PGCs were transcriptionally similar to their in ovo counterparts, with differences in cell proliferation pathways. Our analysis also revealed sex-specific transcriptome differences between male and female cultured PGCs, with notable differences in Smad7 and NCAM2 expression. A comparison of chicken PGCs with pluripotent and somatic cell types identified a set of genes that are exclusive to germ cells, enriched in the germplasm, and associated with germ cell development.
Collapse
Affiliation(s)
- Dadakhalandar Doddamani
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Edinburgh EH25 9RG, UK
| | - Mark Woodcock
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Edinburgh EH25 9RG, UK
| | - Lorna Taylor
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Edinburgh EH25 9RG, UK
| | - Sunil Nandi
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Edinburgh EH25 9RG, UK
| | - Lynn McTeir
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Edinburgh EH25 9RG, UK
| | - Megan G Davey
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Edinburgh EH25 9RG, UK
| | - Jacqueline Smith
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Edinburgh EH25 9RG, UK
| | - Mike J McGrew
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Edinburgh EH25 9RG, UK
| |
Collapse
|
20
|
Kim YM, Woo SJ, Han JY. Strategies for the Generation of Gene Modified Avian Models: Advancement in Avian Germline Transmission, Genome Editing, and Applications. Genes (Basel) 2023; 14:genes14040899. [PMID: 37107658 PMCID: PMC10137648 DOI: 10.3390/genes14040899] [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: 02/24/2023] [Revised: 04/02/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Avian models are valuable for studies of development and reproduction and have important implications for food production. Rapid advances in genome-editing technologies have enabled the establishment of avian species as unique agricultural, industrial, disease-resistant, and pharmaceutical models. The direct introduction of genome-editing tools, such as the clustered regularly interspaced short palindromic repeats (CRISPR) system, into early embryos has been achieved in various animal taxa. However, in birds, the introduction of the CRISPR system into primordial germ cells (PGCs), a germline-competent stem cell, is considered a much more reliable approach for the development of genome-edited models. After genome editing, PGCs are transplanted into the embryo to establish germline chimera, which are crossed to produce genome-edited birds. In addition, various methods, including delivery by liposomal and viral vectors, have been employed for gene editing in vivo. Genome-edited birds have wide applications in bio-pharmaceutical production and as models for disease resistance and biological research. In conclusion, the application of the CRISPR system to avian PGCs is an efficient approach for the production of genome-edited birds and transgenic avian models.
Collapse
Affiliation(s)
| | - Seung-Je Woo
- Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jae-Yong Han
- Avinnogen Co., Ltd., Seoul 08826, Republic of Korea
- Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, College of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
21
|
Han JY, Lee HJ. Genome Editing Mediated by Primordial Germ Cell in Chicken. Methods Mol Biol 2023; 2637:301-312. [PMID: 36773156 DOI: 10.1007/978-1-0716-3016-7_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Genome editing technology has facilitated the studies on exploring specific gene functions in diverse living organisms. The technology has also contributed to creating high-value livestock in industry fields in terms of enhancing productivity or acquiring disease resistance. Particularly, applying genome editing technologies in avian species has been emphasized in both academic and industrial fields due to their unique developmental patterns as well as application possibilities. To accomplish genome editing in avian species, gene integration into chicken primordial germ cell (PGC) genome using a virus or transposition systems has been widely used, and recently developed programmable genome editing technologies including clustered regularly interspaced short palindromic repeat (CRISPR) and CRISPR-associated (Cas9) systems enable to edit the genetic information precisely for maximizing the application potentials of avian species. In these regards, this chapter will cover the methods for producing genome-edited chickens, particularly by CRISPR/Cas9 technologies allowing targeted gene insertion, gene knockout, and gene tagging.
Collapse
Affiliation(s)
- Jae Yong Han
- Department of Agricultural Biotechnology, College of Agriculture and Life Sciences, Seoul National University, Seoul, South Korea.
| | - Hong Jo Lee
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| |
Collapse
|
22
|
Atsuta Y, Suzuki K, Iikawa H, Yaguchi H, Saito D. Prime editing in chicken fibroblasts and primordial germ cells. Dev Growth Differ 2022; 64:548-557. [PMID: 36374008 DOI: 10.1111/dgd.12823] [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: 06/01/2022] [Revised: 09/28/2022] [Accepted: 11/01/2022] [Indexed: 11/16/2022]
Abstract
CRISPR/Cas9-based genome editing technologies are revolutionizing developmental biology. One of the advanced CRISPR-based techniques is prime editing (PE), which enables precise gene modification in multiple model organisms. However, there has been no report of taking advantage of the PE system for gene editing in primordial germ cells (PGCs) thus far. In the current study, we describe a method to apply PE to the genome of chicken fibroblasts and PGCs. By combining PE with a transposon-mediated genomic integration, drug selection, and the single-cell culture method, we successfully generated prime-edited chicken fibroblasts and PGCs. The chicken PGC is widely used as an experimental model to study germ cell formation and as a vector for gene transfer to produce transgenic chickens. Such experimental models are useful in the developmental biology field and as potential bioreactors to produce pharmaceutical and nutritious proteins. Thus, the method presented here will provide not only a powerful tool to investigate gene function in germ cell development but also a basis for generating prime-edited transgenic birds.
Collapse
Affiliation(s)
- Yuji Atsuta
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Japan
| | - Katsuya Suzuki
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Japan
| | - Hiroko Iikawa
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Japan
| | - Haruna Yaguchi
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Japan
| | - Daisuke Saito
- Department of Biology, Faculty of Science, Kyushu University, Fukuoka, Japan
| |
Collapse
|
23
|
Sun Y, Li Y, Zong Y, Mehaisen GMK, Chen J. Poultry genetic heritage cryopreservation and reconstruction: advancement and future challenges. J Anim Sci Biotechnol 2022; 13:115. [PMID: 36210477 PMCID: PMC9549680 DOI: 10.1186/s40104-022-00768-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 08/10/2022] [Indexed: 11/10/2022] Open
Abstract
Poultry genetics resources, including commercial selected lines, indigenous breeds, and experimental lines, are now being irreversibly lost at an alarming rate due to multiple reasons, which further threats the future livelihood and academic purpose. Collections of germplasm may reduce the risk of catastrophic loss of genetic diversity by guaranteeing that a pool of genetic variability is available to ensure the reintroduction and replenishment of the genetic stocks. The setting up of biobanks for poultry is challenging because the high sensitiveness of spermatozoa to freezing–thawing process, inability to cryopreserve the egg or embryo, coupled with the females being heterogametic sex. The progress in cryobiology and biotechnologies have made possible the extension of the range of germplasm for poultry species available in cryobanks, including semen, primordial germ cells, somatic cells and gonads. In this review, we introduce the state-of-the-art technologies for avian genetic resource conservation and breed reconstruction, and discuss the potential challenges for future study and further extending of these technologies to ongoing and future conservation efforts.
Collapse
|
24
|
Establishment of the Primary Avian Gonadal Somatic Cell Lines for Cytogenetic Studies. Animals (Basel) 2022; 12:ani12131724. [PMID: 35804624 PMCID: PMC9264790 DOI: 10.3390/ani12131724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 06/29/2022] [Accepted: 07/02/2022] [Indexed: 12/01/2022] Open
Abstract
Simple Summary We developed a simple method for primary somatic cell culture establishment from the ovaries of the great tits and testes of ten Passerine species. The ovary-derived cell cultures were cultivated until the tenth passage without any noticeable decrease in their proliferative activity, while testis-derived cell cultures demonstrated a decreased proliferation potential. However, sufficient material was available from both cell cultures originating from the ovary and testis to make excellent mitotic metaphase chromosomal preparations. We demonstrated the high efficiency of electroporation for genetic modification of the ovary-derived cell line. Thus, the established ovary-derived cell line could be efficiently used in cytogenetic and genomic studies. Abstract The last decade was marked by a steep rise in avian studies at genomic and cellular levels. Cell lines are important tools for in vitro studies in cell biology and cytogenetics. We developed a simple method of primary somatic cell culture establishment from the ovaries of the great tits (Parus major) and testes of ten Passerine species, characterized the cellular composition of the ovary-derived lines using RT-PCR and immunolocalization of the tissue-specific markers and tested the efficiency of two methods of genetic transformation of the ovary-derived cell line. We found that the ovary-derived cell cultures of the great tit were composed of fibroblasts mainly, but also contained interstitial and granulosa cells. They were cultivated until the 10th passage without any noticeable decrease in their proliferative activity. The testis-derived cell cultures had lower proliferative potential. However, both ovary- and testis-derived cell cultures provided enough material for high quality mitotic metaphase chromosome preparations. The efficiency of its transduction with lentivirus containing a GFP reporter was very low, while electroporation with episomal vectors expressing GFP resulted in a high yield of GFP-positive cells. The proposed method could be used for the generation of high quality material for various cytogenetic and genomic studies.
Collapse
|
25
|
Barkova OY, Larkina TA, Krutikova AA, Polteva EA, Shcherbakov YS, Peglivanyan GK, Pozovnikova MV. Innovative Approaches to Genome Editing in Chickens. CYTOL GENET+ 2022. [DOI: 10.3103/s0095452722020037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
26
|
Hu T, Taylor L, Sherman A, Keambou Tiambo C, Kemp SJ, Whitelaw B, Hawken RJ, Djikeng A, McGrew MJ. A low-tech, cost-effective and efficient method for safeguarding genetic diversity by direct cryopreservation of poultry embryonic reproductive cells. eLife 2022; 11:74036. [PMID: 35074046 PMCID: PMC8789256 DOI: 10.7554/elife.74036] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 01/06/2022] [Indexed: 12/18/2022] Open
Abstract
Chickens are an important resource for smallholder farmers who raise locally adapted, genetically distinct breeds for eggs and meat. The development of efficient reproductive technologies to conserve and regenerate chicken breeds safeguards existing biodiversity and secures poultry genetic resources for climate resilience, biosecurity, and future food production. The majority of the over 1600 breeds of chicken are raised in low and lower to middle income countries under resource-limited, small-scale production systems, which necessitates a low-tech, cost-effective means of conserving diversity is needed. Here, we validate a simple biobanking technique using cryopreserved embryonic chicken gonads. The gonads are quickly isolated, visually sexed, pooled by sex, and cryopreserved. Subsequently, the stored material is thawed and dissociated before injection into sterile host chicken embryos. By using pooled GFP and RFP-labelled donor gonadal cells and Sire Dam Surrogate mating, we demonstrate that chicks deriving entirely from male and female donor germ cells are hatched. This technology will enable ongoing efforts to conserve chicken genetic diversity for both commercial and smallholder farmers, and to preserve existing genetic resources at poultry research facilities.
Collapse
Affiliation(s)
- Tuanjun Hu
- Centre for Tropical Livestock Genetics and Health (CTLGH), The Roslin Institute, University of Edinburgh, Easter Bush Campus
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus
| | - Lorna Taylor
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus
| | - Adrian Sherman
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus
| | - Christian Keambou Tiambo
- Centre for Tropical Livestock Genetics and Health (CTLGH), International Livestock Research Institute (ILRI)
| | - Steven J Kemp
- Centre for Tropical Livestock Genetics and Health (CTLGH), International Livestock Research Institute (ILRI)
| | - Bruce Whitelaw
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus
| | | | - Appolinaire Djikeng
- Centre for Tropical Livestock Genetics and Health (CTLGH), The Roslin Institute, University of Edinburgh, Easter Bush Campus
| | - Michael J McGrew
- Centre for Tropical Livestock Genetics and Health (CTLGH), The Roslin Institute, University of Edinburgh, Easter Bush Campus
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus
| |
Collapse
|
27
|
Ballantyne M, Taylor L, Hu T, Meunier D, Nandi S, Sherman A, Flack B, Henshall JM, Hawken RJ, McGrew MJ. Avian Primordial Germ Cells Are Bipotent for Male or Female Gametogenesis. Front Cell Dev Biol 2021; 9:726827. [PMID: 34660583 PMCID: PMC8511492 DOI: 10.3389/fcell.2021.726827] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/30/2021] [Indexed: 11/13/2022] Open
Abstract
In birds, males are the homogametic sex (ZZ) and females are the heterogametic sex (ZW). Here, we investigate the role of chromosomal sex and germ cell competition on avian germ cell differentiation. We recently developed genetically sterile layer cockerels and hens for use as surrogate hosts for primordial germ cell (PGC) transplantation. Using in vitro propagated and cryopreserved PGCs from a pedigree Silkie broiler breed, we now demonstrate that sterile surrogate layer hosts injected with same sex PGCs have normal fertility and produced pure breed Silkie broiler offspring when directly mated to each other in Sire Dam Surrogate mating. We found that female sterile hosts carrying chromosomally male (ZZ) PGCs formed functional oocytes and eggs, which gave rise to 100% male offspring after fertilization. Unexpectedly, we also observed that chromosomally female (ZW) PGCs carried by male sterile hosts formed functional spermatozoa and produced viable offspring. These findings demonstrate that avian PGCs are not sexually restricted for functional gamete formation and provide new insights for the cryopreservation of poultry and other bird species using diploid stage germ cells.
Collapse
Affiliation(s)
- Maeve Ballantyne
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom.,Centre for Tropical Livestock Genetics and Health, University of Edinburgh, Edinburgh, United Kingdom
| | - Lorna Taylor
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Tuanjun Hu
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom.,Centre for Tropical Livestock Genetics and Health, University of Edinburgh, Edinburgh, United Kingdom
| | - Dominique Meunier
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Sunil Nandi
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Adrian Sherman
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | | | | | | | - Mike J McGrew
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom.,Centre for Tropical Livestock Genetics and Health, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
28
|
Protein expression reveals a molecular sexual identity of avian primordial germ cells at pre-gonadal stages. Sci Rep 2021; 11:19236. [PMID: 34584135 PMCID: PMC8478952 DOI: 10.1038/s41598-021-98454-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 09/06/2021] [Indexed: 11/11/2022] Open
Abstract
In poultry, in vitro propagated primordial germ cells (PGCs) represent an important tool for the cryopreservation of avian genetic resources. However, several studies have highlighted sexual differences exhibited by PGCs during in vitro propagation, which may compromise their reproductive capacities. To understand this phenomenon, we compared the proteome of pregonadal migratory male (ZZ) and female (ZW) chicken PGCs propagated in vitro by quantitative proteomic analysis using a GeLC-MS/MS strategy. Many proteins were found to be differentially abundant in chicken male and female PGCs indicating their early sexual identity. Many of the proteins more highly expressed in male PGCs were encoded by genes localised to the Z sex chromosome. This suggests that the known lack of dosage compensation of the transcription of Z-linked genes between sexes persists at the protein level in PGCs, and that this may be a key factor of their autonomous sex differentiation. We also found that globally, protein differences do not closely correlate with transcript differences indicating a selective translational mechanism in PGCs. Male and female PGC expressed protein sets were associated with differential biological processes and contained proteins known to be biologically relevant for male and female germ cell development, respectively. We also discovered that female PGCs have a higher capacity to uptake proteins from the cell culture medium than male PGCs. This study presents the first evidence of an early predetermined sex specific cell fate of chicken PGCs and their sexual molecular specificities which will enable the development of more precise sex-specific in vitro culture conditions for the preservation of avian genetic resources.
Collapse
|
29
|
Altgilbers S, Klein S, Dierks C, Weigend S, Kues WA. Cultivation and characterization of primordial germ cells from blue layer hybrids (Araucana crossbreeds) and generation of germline chimeric chickens. Sci Rep 2021; 11:12923. [PMID: 34155221 PMCID: PMC8217269 DOI: 10.1038/s41598-021-91490-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/21/2021] [Indexed: 11/29/2022] Open
Abstract
The chicken (Gallus gallus) is one of the most common and widespread domestic species, with an estimated total population of 25 billion birds worldwide. The vast majority of chickens in agriculture originate from hybrid breeding programs and is concentrated on few commercially used high performance lines, whereas numerous local and indigenous breeds are at risk to become extinct. To preserve the genomic resources of rare and endangered chicken breeds innovative methods are necessary. Here, we established a solid workflow for the derivation and biobanking of chicken primordial germ cells (PGCs) from blue layer hybrids. To achieve this, embryos of a cross of heterozygous blue egg layers were sampled to obtain blood derived and gonadal male as well as female PGCs of different genotypes (homozygous, heterozygous and nullizygous blue-allele bearing). The total efficiency of established PGC lines was 45% (47/104) within an average of 49 days until they reached sufficient numbers of cells for cryopreservation. The stem-cell character of the cultivated PGCs was confirmed by SSEA-1 immunostaining, and RT-PCR amplification of the pluripotency- and PGC-specific genes cPOUV, cNANOG, cDAZL and CVH. The Sleeping Beauty transposon system allowed to generate a stable integration of a Venus fluorophore reporter into the chicken genome. Finally, we demonstrated that, after re-transfer into chicken embryos, Venus-positive PGCs migrated and colonized the forming gonads. Semen samples of 13 raised cell chimeric roosters were analyzed by flow cytometry for the efficiency of germline colonization by the transferred PGCs carrying the Venus reporter and their proper differentiation into vital spermatids. Thus, we provide a proof-of-concept study for the potential use of PGCs for the cryobanking of rare breeds or rare alleles.
Collapse
Affiliation(s)
- Stefanie Altgilbers
- Department of Biotechnology, Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, 31535, Neustadt, Germany
| | - Sabine Klein
- Department of Biotechnology, Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, 31535, Neustadt, Germany
| | - Claudia Dierks
- Department of Genetic Ressources, Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, 31535, Neustadt, Germany
| | - Steffen Weigend
- Department of Genetic Ressources, Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, 31535, Neustadt, Germany
| | - Wilfried A Kues
- Department of Biotechnology, Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, 31535, Neustadt, Germany.
| |
Collapse
|
30
|
Zhao R, Zuo Q, Yuan X, Jin K, Jin J, Ding Y, Zhang C, Li T, Jiang J, Li J, Zhang M, Shi X, Sun H, Zhang Y, Xu Q, Chang G, Zhao Z, Li B, Wu X, Zhang Y, Song J, Chen G, Li B. Production of viable chicken by allogeneic transplantation of primordial germ cells induced from somatic cells. Nat Commun 2021; 12:2989. [PMID: 34017000 PMCID: PMC8138025 DOI: 10.1038/s41467-021-23242-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 04/21/2021] [Indexed: 02/03/2023] Open
Abstract
The allogeneic transplantation of primordial germ cells (PGCs) derived from somatic cells overcomes the limitation of avian cloning. Here, we transdifferentiate chicken embryo fibroblasts (CEFs) from black feathered Langshan chickens to PGCs and transplant them into White Plymouth Rock chicken embryos to produce viable offspring with characteristics inherited from the donor. We express Oct4/Sox2/Nanog/Lin28A (OSNL) to reprogram CEFs to induced pluripotent stem cells (iPSCs), which are further induced to differentiate into PGCs by BMP4/BMP8b/EGF. DNA demethylation, histone acetylation and glycolytic activation elevate the iPSC induction efficiency, while histone acetylation and glycolytic inhibition facilitate PGCs formation. The induced PGCs (iPGCs) are transplanted into the recipients, which are self-crossed to produce 189/509 somatic cells derived chicken with the donor's characteristics. Microsatellite analysis and genome sequencing confirm the inheritance of genetic information from the donor. Thus, we demonstrate the feasibility of avian cloning from somatic cells.
Collapse
Affiliation(s)
- Ruifeng Zhao
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Qisheng Zuo
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Xia Yuan
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Kai Jin
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Jing Jin
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Ying Ding
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Chen Zhang
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Tingting Li
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Jingyi Jiang
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Jiancheng Li
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Ming Zhang
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Xiang Shi
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Hongyan Sun
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Yani Zhang
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Qi Xu
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Guobin Chang
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Zhenhua Zhao
- The Poultry Research Institute of Chinese Academy of Agricultural Sciences, Yangzhou, China
| | - Bing Li
- The Poultry Research Institute of Chinese Academy of Agricultural Sciences, Yangzhou, China
| | - Xinsheng Wu
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Yang Zhang
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Jiuzhou Song
- Department of Animal & Avian Sciences, University of Maryland, College Park, MD, USA
| | - Guohong Chen
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China.
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China.
| | - Bichun Li
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China.
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou University, Yangzhou, China.
| |
Collapse
|
31
|
Lázár B, Molnár M, Sztán N, Végi B, Drobnyák Á, Tóth R, Tokodyné Szabadi N, McGrew MJ, Gócza E, Patakiné Várkonyi E. Successful cryopreservation and regeneration of a partridge colored Hungarian native chicken breed using primordial germ cells. Poult Sci 2021; 100:101207. [PMID: 34242944 PMCID: PMC8271167 DOI: 10.1016/j.psj.2021.101207] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 11/30/2022] Open
Abstract
Primordial germ cells (PGCs) are the precursors of germline cells that generate sperm and ova in adults. Thus, they are promising tools for gene editing and genetic preservation, especially in avian species. In this study, we established stable male and female PGC lines from 6Hungarian indigenous chicken breeds with derivation rates ranging from 37.5 to 50 percent. We characterized the PGCs for expression of the germ cell-specific markers during prolonged culture in vitro. An in vivo colonization test was performed on PGCs from four Hungarian chicken breeds and the colonization rates were between 76 and 100%. Cryopreserved PGCs of the donor breed (Partridge color Hungarian) were injected into Black Transylvanian Naked Neck host embryos to form chimeric progeny that, after backcrossing, would permit reconstitution of the donor breed. For 24 presumptive chimeras 13 were male and 11 were female. In the course of backcrossing, 340 chicks were hatched and 17 of them (5%) were pure Partridge colored. Based on the backcrossing 1 hen and 3 roosters of the 24 presumptive chimeras (16.6%) have proven to be germline chimeras. Therefore, it was proven that the original breed can be recovered from primordial germ cells which are stored in the gene bank. To our knowledge, our study is a first that applied feeder free culturing conditions for both male and female cell lines successfully and used multiple indigenous chicken breeds to create a gene bank representing a region (Carpathian Basin).
Collapse
Affiliation(s)
- Bence Lázár
- National Centre for Biodiversity and Gene Conservation, Institute for Farm Animal Gene Conservation, 200 Isaszegi street, 2100 Gödöllő, Hungary; Hungarian University of Agriculture and Life Sciences, Institute of Genetics and Biotechnology, Animal Biotechnology Department, 4 Szent-Györgyi Albert street, 2100 Gödöllő, Hungary.
| | - Mariann Molnár
- National Centre for Biodiversity and Gene Conservation, Institute for Farm Animal Gene Conservation, 200 Isaszegi street, 2100 Gödöllő, Hungary
| | - Nikoletta Sztán
- National Centre for Biodiversity and Gene Conservation, Institute for Farm Animal Gene Conservation, 200 Isaszegi street, 2100 Gödöllő, Hungary
| | - Barbara Végi
- National Centre for Biodiversity and Gene Conservation, Institute for Farm Animal Gene Conservation, 200 Isaszegi street, 2100 Gödöllő, Hungary
| | - Árpád Drobnyák
- National Centre for Biodiversity and Gene Conservation, Institute for Farm Animal Gene Conservation, 200 Isaszegi street, 2100 Gödöllő, Hungary
| | - Roland Tóth
- Hungarian University of Agriculture and Life Sciences, Institute of Genetics and Biotechnology, Animal Biotechnology Department, 4 Szent-Györgyi Albert street, 2100 Gödöllő, Hungary
| | - Nikolett Tokodyné Szabadi
- Hungarian University of Agriculture and Life Sciences, Institute of Genetics and Biotechnology, Animal Biotechnology Department, 4 Szent-Györgyi Albert street, 2100 Gödöllő, Hungary
| | - Michael J McGrew
- The Roslin Institute and Royal Dick School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, EH25 9RG, Midlothian, UK
| | - Elen Gócza
- Hungarian University of Agriculture and Life Sciences, Institute of Genetics and Biotechnology, Animal Biotechnology Department, 4 Szent-Györgyi Albert street, 2100 Gödöllő, Hungary
| | - Eszter Patakiné Várkonyi
- National Centre for Biodiversity and Gene Conservation, Institute for Farm Animal Gene Conservation, 200 Isaszegi street, 2100 Gödöllő, Hungary
| |
Collapse
|
32
|
Gessara I, Dittrich F, Hertel M, Hildebrand S, Pfeifer A, Frankl-Vilches C, McGrew M, Gahr M. Highly Efficient Genome Modification of Cultured Primordial Germ Cells with Lentiviral Vectors to Generate Transgenic Songbirds. Stem Cell Reports 2021; 16:784-796. [PMID: 33740464 PMCID: PMC8072032 DOI: 10.1016/j.stemcr.2021.02.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 12/13/2022] Open
Abstract
The ability to genetically manipulate organisms has led to significant insights into functional genomics in many species. In birds, manipulation of the genome is hindered by the inaccessibility of the one-cell embryo. During embryonic development, avian primordial germ cells (PGCs) migrate through the bloodstream and reach the gonadal anlage, where they develop into mature germ cells. Here, we explored the use of PGCs to produce transgenic offspring in the zebra finch, which is a major animal model for sexual brain differentiation, vocal learning, and vocal communication. Zebra finch PGCs (zfPGCs) obtained from embryonic blood significantly proliferated when cultured in an optimized culture medium and conserved the expression of germ and stem cell markers. Transduction of cultured zfPGCs with lentiviral vectors was highly efficient, leading to strong expression of the enhanced green fluorescent protein. Transduced zfPGCs were injected into the host embryo and transgenic songbirds were successfully generated.
Collapse
Affiliation(s)
- Ivana Gessara
- Max Planck Institute for Ornithology, Eberhard Gwinner Strasse, 82319 Seewiesen, Germany.
| | - Falk Dittrich
- Max Planck Institute for Ornithology, Eberhard Gwinner Strasse, 82319 Seewiesen, Germany
| | - Moritz Hertel
- Max Planck Institute for Ornithology, Eberhard Gwinner Strasse, 82319 Seewiesen, Germany
| | - Staffan Hildebrand
- Institute of Pharmacology and Toxicology, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | | | - Mike McGrew
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Manfred Gahr
- Max Planck Institute for Ornithology, Eberhard Gwinner Strasse, 82319 Seewiesen, Germany
| |
Collapse
|
33
|
Primordial germ cells isolated from individual embryos of red junglefowl and indigenous pheasants of Thailand. Theriogenology 2021; 165:59-68. [PMID: 33640587 DOI: 10.1016/j.theriogenology.2021.02.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/10/2021] [Accepted: 02/13/2021] [Indexed: 11/20/2022]
Abstract
Interspecific germline chimerism mediated by transplantation of primordial germ cells (PGCs) of wild species to domestic hosts promises the conservation of wild birds. Cryopreservation of avian eggs and embryos is impracticable, and currently only frozen PGCs enable conservation of both the male and female descendants. Purebred offspring have been obtained from germline chimeras of wild avian species, proving the feasibility of such technology. In vitro propagation has been optimized for avian PGCs of domestic species; however, evidence is rather limited for successful isolation as well as long-term culture from a single embryo of wild species. With accelerating biodiversity loss, we have committed to preserving current genetic resources by freezing PGCs isolated from individual embryos in addition to their genetic material. We have devised a reliable protocol for the isolation and proliferation of PGCs from wild fowls in the family Phasianidae that are conserved in captive breeding (red junglefowl, bar-tailed pheasant, kalij pheasant, Siamese fireback pheasant, and silver pheasant). We obtained individual isolates of cultured circulating PGCs (49.7%, 79/155) as well as tissue PGCs (92.9%, 144/155). The specific co-culture conditions of autologous embryonic cells, without additional growth factors, facilitated the proliferation of so-called tissue PGCs (the remaining PGCs in embryonic tissue following blood aspiration). Only circulating PGCs left in blood vessels and of PGCs migrating to developing gonads have been previously reported. However, the present study is the first to report on the harvest of ectopic PGCs. The defined conditions sustained continuous proliferation of tissue PGCs for at least six months and maintained PGC identity following cryopreservation. Cultured tissue PGCs of these wild species were extensively characterized for their expression of the germ cell-specific proteins, chicken vasa homolog (CVH) and deleted in azoospermia-like (DAZL), as well as the ability to colonize chicken embryonic gonads. The novel protocol is practical for generating enough PGCs for cryopreservation, transplantation, and additionally, it enables isolation of PGCs from both blood circulation and embryonic tissue simultaneously. For conservation purposes, this approach is potentially applicable more widely to other non-domestic birds than those in the family Phasianidae that were investigated in the present study.
Collapse
|
34
|
Szczerba A, Kuwana T, Paradowska M, Bednarczyk M. In Vitro Culture of Chicken Circulating and Gonadal Primordial Germ Cells on a Somatic Feeder Layer of Avian Origin. Animals (Basel) 2020; 10:E1769. [PMID: 33007811 PMCID: PMC7600596 DOI: 10.3390/ani10101769] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/21/2020] [Accepted: 09/28/2020] [Indexed: 11/28/2022] Open
Abstract
The present study had two aims: (1) To develop a culture system that imitates a normal physiological environment of primordial germ cells (PGCs). There are two types of PGCs in chicken: Circulating blood (cPGCs) and gonadal (gPGCs). The culture condition must support the proliferation of both cPGCs and gPGCs, without affecting their migratory properties and must be deprived of xenobiotic factors, and (2) to propose an easy-to-train, nonlabeling optical technique for the routine identification of live PGCs. To address the first aim, early chicken embryo's feeder cells were examined instead of using feeder cells from mammalian species. The KAv-1 medium at pH 8.0 with the addition of bFGF (basic fibroblast growth factor) was used instead of a conventional culture medium (pH approximately 7.2). Both cPGCs and gPGCs proliferated in vitro and retained their migratory ability after 2 weeks of culture. The cultivated cPGCs and gPGCs colonized the right and/or left gonads of the recipient male and female embryos. To address the second aim, we demonstrated a simple and rapid method to identify live PGCs as bright cells under darkfield illumination. The PGCs rich in lipid droplets in their cytoplasm highly contrasted with the co-cultured feeder layer and other cell populations in the culture.
Collapse
Affiliation(s)
- Agata Szczerba
- Department of Animal Biotechnology and Genetics, Faculty of Animal Breeding and Biology, UTP University of Science and Technology, Mazowiecka 28, 85-084 Bydgoszcz, Poland; (T.K.); (M.P.); (M.B.)
| | | | | | | |
Collapse
|
35
|
Wittig JG, Münsterberg A. The Chicken as a Model Organism to Study Heart Development. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a037218. [PMID: 31767650 DOI: 10.1101/cshperspect.a037218] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Heart development is a complex process and begins with the long-range migration of cardiac progenitor cells during gastrulation. This culminates in the formation of a simple contractile tube with multiple layers, which undergoes remodeling into a four-chambered heart. During this morphogenesis, additional cell populations become incorporated. It is important to unravel the underlying genetic and cellular mechanisms to be able to identify the embryonic origin of diseases, including congenital malformations, which impair cardiac function and may affect life expectancy or quality. Owing to the evolutionary conservation of development, observations made in nonamniote and amniote vertebrate species allow us to extrapolate to human. This review will focus on the contributions made to a better understanding of heart development through studying avian embryos-mainly the chicken but also quail embryos. We will illustrate the classic and recent approaches used in the avian system, give an overview of the important discoveries made, and summarize the early stages of cardiac development up to the establishment of the four-chambered heart.
Collapse
Affiliation(s)
- Johannes G Wittig
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, United Kingdom
| | - Andrea Münsterberg
- School of Biological Sciences, University of East Anglia, Norwich NR4 7TJ, United Kingdom
| |
Collapse
|
36
|
Jin K, Li D, Jin J, Song J, Zhang Y, Chang G, Chen G, Li B. C1EIP Functions as an Activator of ENO1 to Promote Chicken PGCs Formation via Inhibition of the Notch Signaling Pathway. Front Genet 2020; 11:751. [PMID: 32849782 PMCID: PMC7396672 DOI: 10.3389/fgene.2020.00751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 06/23/2020] [Indexed: 12/14/2022] Open
Abstract
The production of germ cells, especially primordial germ cells (PGCs), is important for avian stem cells and reproduction biology. However, key factors involved in the regulation of PGCs remain unknown. Here, we report a PGC-related marker gene: C1EIP (Chromosome 1 Expression in PGCs), whose activation and expression are regulated by the transcription factor STAT3 (signal transducer and activator of transcription 3), histone acetylation, and promoter methylation. C1EIP regulates PGCs formation by mediating the expression of PGC-associated genes, such as CVH (Chicken Vasa Homologous) and CKIT (Chicken KIT proto-oncogene). C1EIP knockdown during embryonic development reduces PGC generation efficiency both in vitro and in ovo. Conversely, C1EIP overexpression increases the formation efficiency of PGCs. C1EIP encodes a cytoplasmic protein that interacts with ENO1 (Enolase 1) in the cytoplasm, inhibits the Notch signaling pathway, and positively regulates PGC generation. Collectively, our findings demonstrate C1EIP as a novel gene involved in PGC formation, which regulates genes involved in embryonic stem cell differentiation through interaction with ENO1 and subsequent inhibition of the Notch signaling pathway by the impression of Myc (MYC proto-oncogene).
Collapse
Affiliation(s)
- Kai Jin
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Dong Li
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University, Medical School, Nanjing, China
| | - Jing Jin
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Jiuzhou Song
- Animal & Avian Sciences, University of Maryland, College Park, College Park, MD, United States
| | - Yani Zhang
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Guobing Chang
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Guohong Chen
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Bichun Li
- Key Laboratory of Animal Breeding Reproduction and Molecular Design for Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, China.,Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou, China
| |
Collapse
|
37
|
Zinovieva NA, Volkova NA, Bagirov VA. Genome Editing: Current State of Research and Application to Animal Husbandry. APPL BIOCHEM MICRO+ 2019. [DOI: 10.1134/s000368381907007x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
38
|
Investigation of the Guinea fowl and domestic fowl hybrids as potential surrogate hosts for avian cryopreservation programmes. Sci Rep 2019; 9:14284. [PMID: 31582777 PMCID: PMC6776557 DOI: 10.1038/s41598-019-50763-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 09/17/2019] [Indexed: 01/15/2023] Open
Abstract
In the last decade, avian gene preservation research has focused on the use of the early precursors of the reproductive cells, the primordial germ cells (PGCs). This is because avian PGCs have a unique migration route through the vascular system which offers easy accessibility. Furthermore, culturing of the cells in vitro, freezing/thawing, reintegration into a recipient embryo and the development of the germ cells can be carried out in well-defined laboratory circumstances. The efficient recovery of the donor genotype and the frequency of germline transmission from the surrogate host animals are still areas which need further development. Thus, the aim of the present study was to investigate an infertile interspecific hybrid (recipient) as an appropriate host for primordial germ cells from native poultry breeds. Guinea fowl × chicken hybrids were produced, the crossing was repeated inversely. The phenotype, the hatching time, the hatching rate, the sex ratio, the presence of own germ cells, the fertility and the phenotype of viable hybrids and the incidence of chromosomal abnormalities of dead hybrid embryos were described. 6.65% viable offspring was obtained with crossing of Guinea fowl females with domestic fowl males. Crossing of domestic fowl hens with Guinea fowl male resulted in lower fertility, 0.14% viable offspring. Based on the investigations, the observed offspring from the successful crossing were sterile male hybrids, thus an extreme form of Haldane’s rule was manifested. The sterile hybrid male embryos were tested by injecting fluorescently labeled chicken PGCs. The integration rate of labeled PGCs was measured in 7.5-day, 14.5-day and 18.5-day old embryonic gonads. 50%, 5.3% and 2.4% of the injected hybrid embryos survived and 40%, 5.3% and 2.4% of the examined gonads contained fluorescent labeled donor PGCs. Therefore, these sterile hybrid males may be suitable recipients for male PGCs and possibly for female PGCs although with lower efficiency. This research work shows that the sterility of hybrids can be used in gene conservation to be a universal host for PGCs of different avian species.
Collapse
|
39
|
Reviving rare chicken breeds using genetically engineered sterility in surrogate host birds. Proc Natl Acad Sci U S A 2019; 116:20930-20937. [PMID: 31575742 DOI: 10.1073/pnas.1906316116] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In macrolecithal species, cryopreservation of the oocyte and zygote is not possible due to the large size and quantity of lipid deposited within the egg. For birds, this signifies that cryopreserving and regenerating a species from frozen cellular material are currently technically unfeasible. Diploid primordial germ cells (PGCs) are a potential means to freeze down the entire genome and reconstitute an avian species from frozen material. Here, we examine the use of genetically engineered (GE) sterile female layer chicken as surrogate hosts for the transplantation of cryopreserved avian PGCs from rare heritage breeds of chicken. We first amplified PGC numbers in culture before cryopreservation and subsequent transplantation into host GE embryos. We found that all hatched offspring from the chimera GE hens were derived from the donor rare heritage breed broiler PGCs, and using cryopreserved semen, we were able to produce pure offspring. Measurement of the mutation rate of PGCs in culture revealed that 2.7 × 10-10 de novo single-nucleotide variants (SNVs) were generated per cell division, which is comparable with other stem cell lineages. We also found that endogenous avian leukosis virus (ALV) retroviral insertions were not mobilized during in vitro propagation. Taken together, these results show that mutation rates are no higher than normal stem cells, essential if we are to conserve avian breeds. Thus, GE sterile avian surrogate hosts provide a viable platform to conserve and regenerate avian species using cryopreserved PGCs.
Collapse
|
40
|
Establishment and Characterization of a Novel Tissue-specific DNA Construct and Culture System with Potential for Avian Bioreactor Generation. Mol Biotechnol 2019; 61:400-409. [PMID: 30945164 DOI: 10.1007/s12033-019-00170-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Transgenic chickens are of great interest for the production of recombinant proteins in their eggs. However, the use of constitutive strong promoters or the tissue-specific ovalbumin promoter for the generation of the transgenic chickens have different drawbacks that have to be overcome in order to make chicken bioreactor an efficient production system. This prompted us to investigate the use of an alternative tissue-specific promoter, the vitellogenin promoter, which could overcome the difficulties currently found in the generation of chicken bioreactors. In the present work we establish and characterize a DNA construct consisting of a fragment of the 5´-flanking region of the chicken vitellogenin II gene cloned in a reporter vector. This construct is capable of showing the ability of the promoter to drive expression of a reporting gene in a tissue-specific manner and in a way that closely resembles physiologic regulation of vitellogenin, making it an ideal candidate to be used in the future for generation of avian bioreactors. Besides, we validate an in vitro culture system to test the performance of the DNA construct under study that could be used as a practical tool before generating any transgenic chicken. These results are important since they provide the proof of concept for the use of the vitellogenin promoter for future genetic modification of chickens bioreactors with improved characteristics in terms of quality of the recombinant protein produced.
Collapse
|
41
|
Wang S, Wang Y, Shen D, Zhang L, Chen W, Chan S, Guan Z, Song C, Gao B. ZB transposon and chicken vasa homologue (Cvh) promoter interact to increase transfection efficiency of primordial germ cells in vivo. Br Poult Sci 2019; 60:724-728. [PMID: 31267766 DOI: 10.1080/00071668.2019.1639138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
1. In order to increase the efficiency of generating transgenic chicken, this trial focused on two points: primordial germ cells (PGCs)transfection in vivo and a germline-specific promoter.2. In order to transfect PGCs in vivo, two plasmids (pZB-CAG-GFP, pCMV-ZB)were co-injected into chicken embryos via the subgerminal cavity at Hamburger and Hamilton (HH) stage 2-3 or via blood vessel at HH stage 13-14. Results showed that the percentage of GFP+ embryos, viability and hatching rate of embryos injected at HH stage 13-14 were significantly higher than that at HH stage 2-3.3. Two plasmid transposon systems were used for chicken embryo micro-injections. The donor plasmid, with a green fluorescent protein (GFP) reporter gene, was mediated by the ZB transposon. The helper plasmid was a transposase expression vector driven by the promoter of the chicken vasa homologue (Cvh) gene or Human cytomegalovirus (CMV) promoter. Results showed that 60.98% of gonads in Cvh group expressed GFP, which was 52.50% higher than seen in the CMV group. Only gonad tissue from the Cvh group showed any GFP signal, whereas both gonads and other tissues in the CMV group showed green fluorescence.4. The data suggested that ZB transposon-mediated gene transfer was efficient for transfecting PGCs in vivo; the Cvh promoter drove the transposase gene specifically in the germline and increased the efficiency of germline transmission. Blood vessels injection at HH stage 13-14 may be a more efficient route for PGCs transfection in vivo.
Collapse
Affiliation(s)
- S Wang
- College of Animal Science & Technology, Joint International Research Laboratory of Agriculture and Agri-product Safety, Yangzhou University, Yangzhou, Jiangsu, China
| | - Y Wang
- College of Animal Science & Technology, Joint International Research Laboratory of Agriculture and Agri-product Safety, Yangzhou University, Yangzhou, Jiangsu, China
| | - D Shen
- College of Animal Science & Technology, Joint International Research Laboratory of Agriculture and Agri-product Safety, Yangzhou University, Yangzhou, Jiangsu, China
| | - L Zhang
- College of Animal Science & Technology, Joint International Research Laboratory of Agriculture and Agri-product Safety, Yangzhou University, Yangzhou, Jiangsu, China
| | - W Chen
- College of Animal Science & Technology, Joint International Research Laboratory of Agriculture and Agri-product Safety, Yangzhou University, Yangzhou, Jiangsu, China
| | - S Chan
- College of Animal Science & Technology, Joint International Research Laboratory of Agriculture and Agri-product Safety, Yangzhou University, Yangzhou, Jiangsu, China
| | - Z Guan
- College of Animal Science & Technology, Joint International Research Laboratory of Agriculture and Agri-product Safety, Yangzhou University, Yangzhou, Jiangsu, China
| | - C Song
- College of Animal Science & Technology, Joint International Research Laboratory of Agriculture and Agri-product Safety, Yangzhou University, Yangzhou, Jiangsu, China
| | - B Gao
- College of Animal Science & Technology, Joint International Research Laboratory of Agriculture and Agri-product Safety, Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
42
|
Chen YC, Lin SP, Chang YY, Chang WP, Wei LY, Liu HC, Huang JF, Pain B, Wu SC. In vitro culture and characterization of duck primordial germ cells. Poult Sci 2019; 98:1820-1832. [PMID: 30462334 PMCID: PMC6414036 DOI: 10.3382/ps/pey515] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 10/15/2018] [Indexed: 12/15/2022] Open
Abstract
This study aimed to isolate, culture, and characterize duck primordial germ cells (PGCs) and to compare these cells with chicken PGCs. We first cultured Muscovy duck (Cairina moschata) circulating PGCs and gonadal PGCs (gPGCs) in the modified serum-containing medium used to amplify chicken PGCs. gPGCs were found to proliferate better in serum-free chemically defined medium than in serum-containing medium. Thereafter, gPGCs were similarly isolated from 2 other duck breeds, the Pekin duck (Anas platyrhynchos) and the hybrid mule duck (C. moschata × A. platyrhynchos), and amplified for a limited period of time in the chemically defined culture condition, but sufficiently to be characterized and transplanted. Cultured gPGCs of all 3 duck breeds were characterized by Periodic acid-Schiff staining, immunocytochemical staining, and expression analysis of germline-specific and pluripotency genes. Cultured duck gPGCs colonized the gonads after being genetically labeled and injected into recipient embryos. Taken together, these results demonstrate that duck PGCs retain their germline characteristics after being isolated, expanded in vitro, and genetically modified. Further studies are required to establish the optimal conditions for long-term culture of duck PGCs, which may involve supplementing the culture medium with other growth factors or compounds.
Collapse
Affiliation(s)
- Yi-Chen Chen
- Institute of Biotechnology, National Taiwan University, Taipei 10672, Taiwan.,Univ Lyon, Université Lyon 1, Stem Cell and Brain Research Institute, U1208, USC1361, INSERM, INRA, Bron 69500, France
| | - Shau-Ping Lin
- Institute of Biotechnology, National Taiwan University, Taipei 10672, Taiwan.,Agricultural Biotechnology Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Yi-Ying Chang
- Ilan Branch, Livestock Research Institute, Council of Agriculture, Executive Yuan, Ilan 26846, Taiwan
| | - Wei-Peng Chang
- Ilan Branch, Livestock Research Institute, Council of Agriculture, Executive Yuan, Ilan 26846, Taiwan
| | - Liang-Yuan Wei
- Ilan Branch, Livestock Research Institute, Council of Agriculture, Executive Yuan, Ilan 26846, Taiwan
| | - Hsiu-Chou Liu
- Ilan Branch, Livestock Research Institute, Council of Agriculture, Executive Yuan, Ilan 26846, Taiwan
| | - Jeng-Fang Huang
- Ilan Branch, Livestock Research Institute, Council of Agriculture, Executive Yuan, Ilan 26846, Taiwan.,Livestock Research Institute, Council of Agriculture, Executive Yuan, Tainan 71246, Taiwan
| | - Bertrand Pain
- Univ Lyon, Université Lyon 1, Stem Cell and Brain Research Institute, U1208, USC1361, INSERM, INRA, Bron 69500, France
| | - Shinn-Chih Wu
- Institute of Biotechnology, National Taiwan University, Taipei 10672, Taiwan.,Department of Animal Science and Technology, National Taiwan University, Taipei 10672, Taiwan
| |
Collapse
|
43
|
Huss DJ, Saias S, Hamamah S, Singh JM, Wang J, Dave M, Kim J, Eberwine J, Lansford R. Avian Primordial Germ Cells Contribute to and Interact With the Extracellular Matrix During Early Migration. Front Cell Dev Biol 2019; 7:35. [PMID: 30984757 PMCID: PMC6447691 DOI: 10.3389/fcell.2019.00035] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 02/26/2019] [Indexed: 01/10/2023] Open
Abstract
During early avian development, primordial germ cells (PGC) are highly migratory, moving from the central area pellucida of the blastoderm to the anterior extra-embryonic germinal crescent. The PGCs soon move into the forming blood vessels by intravasation and travel in the circulatory system to the genital ridges where they participate in the organogenesis of the gonads. This complex cellular migration takes place in close association with a nascent extracellular matrix that matures in a precise spatio-temporal pattern. We first compiled a list of quail matrisome genes by bioinformatic screening of human matrisome orthologs. Next, we used single cell RNA-seq analysis (scRNAseq) to determine that PGCs express numerous ECM and ECM-associated genes in early embryos. The expression of select ECM transcripts and proteins in PGCs were verified by fluorescent in situ hybridization (FISH) and immunofluorescence (IF). Live imaging of transgenic quail embryos injected with fluorescent antibodies against fibronectin and laminin, showed that germinal crescent PGCs display rapid shape changes and morphological properties such as blebbing and filopodia while surrounded by, or in close contact with, an ECM fibril meshwork that is itself in constant motion. Injection of anti-β1 integrin CSAT antibodies resulted in a reduction of mature fibronectin and laminin fibril meshwork in the germinal crescent at HH4-5 but did not alter the active motility of the PGCs or their ability to populate the germinal crescent. These results suggest that integrin β1 receptors are important, but not required, for PGCs to successfully migrate during embryonic development, but instead play a vital role in ECM fibrillogenesis and assembly.
Collapse
Affiliation(s)
- David J. Huss
- Department of Radiology, Children's Hospital Los Angeles, Los Angeles, CA, United States
- Translational Imaging Center, University of Southern California, Los Angeles, CA, United States
| | - Sasha Saias
- Department of Radiology, Children's Hospital Los Angeles, Los Angeles, CA, United States
| | - Sevag Hamamah
- Department of Radiology, Children's Hospital Los Angeles, Los Angeles, CA, United States
| | - Jennifer M. Singh
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, United States
- Penn Genome Frontiers Institute, University of Pennsylvania, Philadelphia, PA, United States
| | - Jinhui Wang
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, United States
- Penn Genome Frontiers Institute, University of Pennsylvania, Philadelphia, PA, United States
| | - Mohit Dave
- Department of Radiology, Children's Hospital Los Angeles, Los Angeles, CA, United States
| | - Junhyong Kim
- Penn Genome Frontiers Institute, University of Pennsylvania, Philadelphia, PA, United States
- Department of Biology, University of Pennsylvania, Philadelphia, PA, United States
| | - James Eberwine
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, United States
- Penn Genome Frontiers Institute, University of Pennsylvania, Philadelphia, PA, United States
| | - Rusty Lansford
- Department of Radiology, Children's Hospital Los Angeles, Los Angeles, CA, United States
- Translational Imaging Center, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
44
|
Yu F, Zhu Z, Chen X, Huang J, Jia R, Pan J. Isolation, characterization and germline chimera preparation of primordial germ cells from the Chinese Meiling chicken. Poult Sci 2019; 98:566-572. [DOI: 10.3382/ps/pey410] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 08/10/2018] [Indexed: 11/20/2022] Open
|
45
|
Kheirabadi M, Nabipour A, Dehghani H, Behnam-Rasuli M. Ultrastructure of ovarian germ cells in the ostrich (Struthio camelus) embryo. BULGARIAN JOURNAL OF VETERINARY MEDICINE 2019. [DOI: 10.15547/bjvm.2086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In this study, the ultrastructural development of germ cells in the ostrich embryo was analysed. The nuclear organisation and morphological characteristics of cytoplasm in the developing germ cells, on embryonic days 20, 26, and 36 and the day of hatching (5 samples from each stage) was analysed using transmission electron microscopy (TEM). Germ cells located in the cortex of left ovaries were identified by their large size and centrally located nucleus, with a conspicuous nucleolus. In these cells, the cytoplasm contained an abundance of mitochondria and free ribosomes. The structure of Balbiani body, a villous-like elevation in wide intercellular space and desmosome junction between two adjacent germ cells was also studied. The germ cells during embryonic development showed structural differences in both the nucleus and cytoplasm.
Collapse
|
46
|
Three-dimensional culture of chicken primordial germ cells (cPGCs) in defined media containing the functional polymer FP003. PLoS One 2018; 13:e0200515. [PMID: 30240390 PMCID: PMC6150485 DOI: 10.1371/journal.pone.0200515] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Accepted: 08/30/2018] [Indexed: 11/29/2022] Open
Abstract
Scalable production of avian cell lines exhibits a valuable potential on therapeutic application by producing recombinant proteins and as the substrate for virus growth due to the special glycosylation occurs in avian species. Chicken primordial germ cells (cPGCs), a germinal pluripotent avian cell type, present the ability of self-renewal, an anchorage-independent cell growth and the ability to be genetically modified. This cell type could be an interesting bioreactor system for industrial purposes. This study sought to establish an expandable culture system with defined components for three-dimensional (3D) culture of cPGCs. cPGCs were cultured in medium supplemented with the functional polymer FP003. Viscoelasticity was low in this medium but cPGCs did not sediment in culture and efficiencies of space and nutrient utilization were thus enhanced and consequently their expansion was improved. The total number of cPGCs increased by 17-fold after 1 week of culture in 3D-FAot medium, an aseric defined medium containing FP003 polymer, FGF2 and Activin A as growth factors and Ovotransferrin as protein. Moreover, cPGC cell lines stably expressed the germline-specific reporter VASA:tdTOMATO, as well as other markers of cPGCs, for more than 1 month upon culture in 3D-FAot medium, indicating that the characteristics of these cells are maintained. In summary, this novel 3D culture system can be used to efficiently expand cPGCs in suspension without mechanical stirring, which is available for long-term culture and no loss of cellular properties was found. This system provides a platform for large-scale culture of cPGCs.
Collapse
|
47
|
Derivation of chicken primordial germ cells using an indirect Co-culture system. Theriogenology 2018; 123:83-89. [PMID: 30292859 DOI: 10.1016/j.theriogenology.2018.09.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 09/14/2018] [Accepted: 09/14/2018] [Indexed: 11/23/2022]
Abstract
Primordial germ cells (PGCs) are promising genetic resources for avian studies including modified animals. However, chicken PGCs are slow to proliferate and gradually lose germline competency after long-term culture, which hinders their application in avian biotechnology. Thus, we developed a robust method for the isolation and rapid propagation of PGCs using an indirect co-culture system. PGCs derived from a pair of embryonic chicken gonads were expanded to 1 × 106 within 2 weeks, and no sex bias was observed in. These PGCs presented high capacity of germline transmission and produced donor-derived offspring after injection into the chicken embryos. This system allows the efficient gene-banking of chicken species and can facilitate the production of chickens bearing a desired phenotype via genomic editing.
Collapse
|
48
|
Oishi I, Yoshii K, Miyahara D, Tagami T. Efficient production of human interferon beta in the white of eggs from ovalbumin gene-targeted hens. Sci Rep 2018; 8:10203. [PMID: 29976933 PMCID: PMC6033876 DOI: 10.1038/s41598-018-28438-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 06/22/2018] [Indexed: 12/20/2022] Open
Abstract
Transgenic chickens could potentially serve as bioreactors for commercial production of recombinant proteins in egg white. Many transgenic chickens have been generated by randomly integrating viral vectors into their genomes, but transgene expression has proved insufficient and/or limited to the initial cohort. Herein, we demonstrate the feasibility of integrating human interferon beta (hIFN-β) into the chicken ovalbumin locus and producing hIFN-β in egg white. We knocked in hIFN-β into primordial germ cells using a CRISPR/Cas9 protocol and then generated germline chimeric roosters by cell transplantation into recipient embryos. Two generation-zero founder roosters produced hIFN-β knock-in offspring, and all knock-in female offspring produced abundant egg-white hIFN-β (~3.5 mg/ml). Although female offspring of the first generation were sterile, their male counterparts were fertile and produced a second generation of knock-in hens, for which egg-white hIFN-β production was comparable with that of the first generation. The hIFN-β bioactivity represented only ~5% of total egg-white hIFN-β, but unfolding and refolding of hIFN-β in the egg white fully recovered the bioactivity. These results suggest that transgene insertion at the chicken ovalbumin locus can result in abundant and stable expression of an exogenous protein deposited into egg white and should be amenable to industrial applications.
Collapse
Affiliation(s)
- Isao Oishi
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, 1-8-31, Midorioka, Ikeda, Osaka, 563-8577, Japan.
| | - Kyoko Yoshii
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, 1-8-31, Midorioka, Ikeda, Osaka, 563-8577, Japan
| | - Daichi Miyahara
- Animal Breeding and Reproduction Research Division, National Agriculture and Food Research Organization, Institute of Livestock and Grassland Science, 2 Ikenodai, Tsukuba, Ibaraki, 305-0901, Japan
| | - Takahiro Tagami
- Animal Breeding and Reproduction Research Division, National Agriculture and Food Research Organization, Institute of Livestock and Grassland Science, 2 Ikenodai, Tsukuba, Ibaraki, 305-0901, Japan
| |
Collapse
|
49
|
Abstract
Primordial germ cells (PGCs), precursors of functional gametes, offer great promise for the use of genetic resources and transgenesis in chickens. PGCs can be isolated from the developing embryo at diverse early stages and are subsequently expandable in vitro. In vitro proliferating chicken PGCs can facilitate the production of efficient germline chimeras and transgenic chickens. Here, we describe methods to isolate and characterize PGCs from chicken embryos and their application to transgenesis.
Collapse
|
50
|
Long-term in vitro culture and preliminary establishment of chicken primordial germ cell lines. PLoS One 2018; 13:e0196459. [PMID: 29709001 PMCID: PMC5927411 DOI: 10.1371/journal.pone.0196459] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 04/15/2018] [Indexed: 11/19/2022] Open
Abstract
Primordial germ cells (PGCs) are precursors of functional gametes and can be used as efficient transgenic tools and carriers in bioreactors. Few methods for long-term culture of PGCs are available. In this study, we tested various culture conditions for PGCs, and used the optimum culture system to culture chicken gonad PGCs for about three hundred days. Long-term-cultured PGCs were detected and characterized by karyotype analysis, immunocytochemical staining of SSEA-1, c-kit, Sox2, cDAZL, and quantitative RT-PCR for specific genes like Tert, DAZL, POUV, and NANOG. Cultured PGCs labeled with PKH26 were reinjected into Stage X recipient embryos and into the dorsal aorta of Stage 14–17 embryos to assay their ability of migration into the germinal crescent and gonads, respectively. In conclusion, the most suitable culture system for PGCs is as follows: feeder layer cells treated with 20 μg/mL mitomycin C for 2 hours, and with 50% conditioned medium added to the factor culture medium. PGCs cultured in this system retain their pluripotency and the unique ability of migration without transformation, indicating the successful preliminary establishment of chicken primordial germ cell lines and these PGCs can be considered for use as carriers in transgenic bioreactors.
Collapse
|