1
|
Meng S, Miao A, Wu S, Du X, Gao F. Genetically modified chickens as bioreactors for protein-based drugs. Front Genome Ed 2025; 6:1522837. [PMID: 39845893 PMCID: PMC11753250 DOI: 10.3389/fgeed.2024.1522837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 12/18/2024] [Indexed: 01/24/2025] Open
Abstract
Protein drug production encompasses various methods, among which animal bioreactors are emerging as a transgenic system. Animal bioreactors have the potential to reduce production costs and increase efficiency, thereby producing recombinant proteins that are crucial for therapeutic applications. Various species, including goats, cattle, rabbits, and poultry, have been genetically engineered to serve as bioreactors. This review delves into the analysis and comparison of different expression systems for protein drug production, highlighting the advantages and limitations of microbial, yeast, plant cell, and mammalian cell expression systems. Additionally, the emerging significance of genetically modified chickens as a potential bioreactor system for producing protein-based drugs is highlighted. The avian bioreactor enables the expression of target genes in ovarian cells, resulting in the production of corresponding gene expression products in egg whites. This production method boasts advantages such as a short cycle, high production efficiency, low research costs, and the expression products being closer to their natural state and easier to purify. It demonstrates immense potential in production applications, scientific research, and sustainable development. The utilization of advanced gene editing technologies, such as CRISPR/Cas9, has revolutionized the precision and efficiency of generating genetically modified chickens. This has paved the way for enhanced production of recombinant therapeutic proteins with desired glycosylation patterns and reduced immunogenic responses.
Collapse
Affiliation(s)
- Shujuan Meng
- Frontiers Science Center for Molecular Design Breeding (MOE), State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Aijun Miao
- Frontiers Science Center for Molecular Design Breeding (MOE), State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Sen Wu
- Frontiers Science Center for Molecular Design Breeding (MOE), State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
- Sanya Institute of China Agricultural University, Sanya, China
| | - Xuguang Du
- Frontiers Science Center for Molecular Design Breeding (MOE), State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
- Sanya Institute of China Agricultural University, Sanya, China
| | - Fei Gao
- Frontiers Science Center for Molecular Design Breeding (MOE), State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| |
Collapse
|
2
|
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
|
3
|
Wattad NR, Ozer E, Altgilbers S, Klein C, Cohen E, Zuckrman O, Sessler E, Hadad T, Alcalay Y, Abdu U. Intratesticular versus intraperitoneal Busulfan administration: a comparative study on spermatogenesis suppression in quails and chickens. Poult Sci 2024; 103:103890. [PMID: 38870613 PMCID: PMC11225892 DOI: 10.1016/j.psj.2024.103890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/15/2024] [Accepted: 05/20/2024] [Indexed: 06/15/2024] Open
Abstract
Generation of transgenic birds can be achieved by temporal suppression of endogenous spermatogenesis in males prior to primordial germ cell implantation. One of many established methods to induce male sterility is the intraperitoneal injection of busulfan, an alkylating agent. Nevertheless, the use of busulfan injections, which may also affect hematopoietic stem cells, carries the risk of potential lethality in animals. Given their safety and non-toxic nature, it has been demonstrated that intratesticular busulfan injections in mammals are less effective than intraperitoneal injections. This study aimed to compare, for the first time, the sterility and toxicity effects of intraperitoneal vs. intratesticular busulfan injections in quail and chickens. Our experimental design involved a previously established single intraperitoneal busulfan injection of 40 mg/kg of body weight (BW). In quail, busulfan was then administered intratesticularly at 3 different concentrations (6, 12, and 20 mg/kg BW), while in chickens, the working concentration was 20 mg/kg BW. We found that a single intraperitoneal busulfan injection of 40 mg/kg of BW resulted in 100% mortality in the treated roosters. In quails, however, this concentration only caused a temporary suppression of fertility for a 15-d period. Moreover, we found that a higher dose of intratesticular injection of busulfan is required to suppress spermatogenesis in quail (20 mg/kg BW) compared to mammals (4 mg/kg BW). Following these findings, we further confirmed that intratesticular injection of 20 mg/kg BW busulfan into roosters did not affect their overall viability. However, it induced a temporary state of male sterility, consistent with the effects observed with intraperitoneal injections. Hence, our data demonstrate that quail and chicken respond differently to busulfan administration. Furthermore, the present study provides evidence that direct injection into the rooster testes causes less physiological stress than intraperitoneal injection.
Collapse
Affiliation(s)
- Noor R Wattad
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Eden Ozer
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Stefanie Altgilbers
- Friedrich-Loeffler-Institut - Federal Research Institute for Animal Health (FLI), Institute of Farm Animal Genetics, Department of Biotechnology, Neustadt 31535, Germany
| | - Claudia Klein
- Friedrich-Loeffler-Institut - Federal Research Institute for Animal Health (FLI), Institute of Farm Animal Genetics, Department of Biotechnology, Neustadt 31535, Germany
| | - Eyal Cohen
- COPIA Agro & Food, Herzliya, 4672501, Israel
| | | | | | - Tamar Hadad
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | | | - Uri Abdu
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel.
| |
Collapse
|
4
|
Guillot C, Djeffal Y, Serra M, Pourquié O. Control of epiblast cell fate by mechanical cues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.24.600402. [PMID: 38979228 PMCID: PMC11230242 DOI: 10.1101/2024.06.24.600402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
In amniotes, embryonic tissues originate from multipotent epiblast cells, arranged in a mosaic of presumptive territories. How these domains fated to specific lineages become segregated during body formation remains poorly understood. Using single cell RNA sequencing analysis and lineage tracing in the chicken embryo, we identify epiblast cells contributing descendants to the neural tube, somites and lateral plate after completion of gastrulation. We show that intercalation after cell division generates important movements of epiblast cells which lead to their relocation to different presumptive territories, explaining this broad spectrum of fates. This tissue remodeling phase is transient, being soon followed by the establishment of boundaries restricting cell movements therefore defining the presumptive territories of the epiblast. Finally, we find that the epiblast faces distinct mechanical constraints along the antero-posterior axis, leading to cell fate alterations when challenged. Together, we demonstrate the critical role of mechanical cues in epiblast fate determination.
Collapse
|
5
|
Wu M, Maiorano G, Stadnicka K. Protein profiles in the transfected oviductal secreting cells of laying hen (Gallus gallus domesticus). Poult Sci 2024; 103:103305. [PMID: 38198917 PMCID: PMC10792652 DOI: 10.1016/j.psj.2023.103305] [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: 08/18/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 01/12/2024] Open
Abstract
Due to the intensive development of novel biopharming applications, there is a need for the in vitro verification models prior to in vivo testing. Laying hen has been already applied as an animal bioreactor to produce the therapeutical enzyme in a rare disease called lysosomal acid lipase deficiency. In this study, we aimed to verify how the proteome of the transfected oviduct epithelial cells would be affected by genetic nonviral modification with the human exogene. The study was based on a previously developed method to cultivate chicken oviduct epithelial cells (COEC). The typical characteristics of the COEC epithelial cells were retained across the experiments. The mean efficiency of nucleofection ranged from 2.6 to 19.7% depending on the cells' isolation and location in the oviduct (upper, infundibulum site, or magnum). The PCR confirmed the incorporation of human interferon alpha2a (hIFNα2a) exogene into the nucleofected COEC but, the production of hIFNα2a protein did not exceed the detection level in this study. The ovalbumin protein was detected in the nontransfected and transfected COEC, which confirmed the normal secreting functions of the cells subject to modification. Proteomic analysis revealed an increase in abundance of the cell adhesion molecules and collagen molecules after introducing gene under ovalbumin promoter. According to the bioinformatic analyses there was a limited negative impact of transfection on cells, and the normal biochemical pathways were not severely disordered. In conclusion, the observations provide new knowledge about the proteomic profile of the manipulated COEC with regard to the retained normal functionality of the cells, which can be informative for avian biopharma research.
Collapse
Affiliation(s)
- Mengjun Wu
- Department of Agricultural, Environmental and Food Sciences, University of Molise, 86100 Campobasso, Italy
| | - Giuseppe Maiorano
- Department of Agricultural, Environmental and Food Sciences, University of Molise, 86100 Campobasso, Italy
| | - Katarzyna Stadnicka
- Faculty of Health Sciences, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-821 Bydgoszcz, Poland.
| |
Collapse
|
6
|
Cooper F, Souilhol C, Haston S, Gray S, Boswell K, Gogolou A, Frith TJR, Stavish D, James BM, Bose D, Kim Dale J, Tsakiridis A. Notch signalling influences cell fate decisions and HOX gene induction in axial progenitors. Development 2024; 151:dev202098. [PMID: 38223992 PMCID: PMC10911136 DOI: 10.1242/dev.202098] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 12/20/2023] [Indexed: 01/16/2024]
Abstract
The generation of the post-cranial embryonic body relies on the coordinated production of spinal cord neurectoderm and presomitic mesoderm cells from neuromesodermal progenitors (NMPs). This process is orchestrated by pro-neural and pro-mesodermal transcription factors that are co-expressed in NMPs together with Hox genes, which are essential for axial allocation of NMP derivatives. NMPs reside in a posterior growth region, which is marked by the expression of Wnt, FGF and Notch signalling components. Although the importance of Wnt and FGF in influencing the induction and differentiation of NMPs is well established, the precise role of Notch remains unclear. Here, we show that the Wnt/FGF-driven induction of NMPs from human embryonic stem cells (hESCs) relies on Notch signalling. Using hESC-derived NMPs and chick embryo grafting, we demonstrate that Notch directs a pro-mesodermal character at the expense of neural fate. We show that Notch also contributes to activation of HOX gene expression in human NMPs, partly in a non-cell-autonomous manner. Finally, we provide evidence that Notch exerts its effects via the establishment of a negative-feedback loop with FGF signalling.
Collapse
Affiliation(s)
- Fay Cooper
- School of Biosciences, The University of Sheffield, Sheffield S10 2TN, UK
- Neuroscience Institute, The University of Sheffield, Sheffield S10 2TN, UK
| | - Celine Souilhol
- School of Biosciences, The University of Sheffield, Sheffield S10 2TN, UK
- Neuroscience Institute, The University of Sheffield, Sheffield S10 2TN, UK
- Biomolecular Sciences Research Centre, Department of Biosciences and Chemistry, Sheffield Hallam University, Sheffield S1 1WB, UK
| | - Scott Haston
- Developmental Biology and Cancer, Birth Defects Research Centre, UCL GOS Institute of Child Health, London WC1N 1EH, UK
- Division of Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee DD1 4HN, UK
| | - Shona Gray
- Division of Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee DD1 4HN, UK
| | - Katy Boswell
- School of Biosciences, The University of Sheffield, Sheffield S10 2TN, UK
- Neuroscience Institute, The University of Sheffield, Sheffield S10 2TN, UK
| | - Antigoni Gogolou
- School of Biosciences, The University of Sheffield, Sheffield S10 2TN, UK
- Neuroscience Institute, The University of Sheffield, Sheffield S10 2TN, UK
| | - Thomas J. R. Frith
- School of Biosciences, The University of Sheffield, Sheffield S10 2TN, UK
- Neuroscience Institute, The University of Sheffield, Sheffield S10 2TN, UK
| | - Dylan Stavish
- School of Biosciences, The University of Sheffield, Sheffield S10 2TN, UK
- Neuroscience Institute, The University of Sheffield, Sheffield S10 2TN, UK
| | - Bethany M. James
- School of Biosciences, The University of Sheffield, Sheffield S10 2TN, UK
- Neuroscience Institute, The University of Sheffield, Sheffield S10 2TN, UK
| | - Daniel Bose
- School of Biosciences, The University of Sheffield, Sheffield S10 2TN, UK
- Neuroscience Institute, The University of Sheffield, Sheffield S10 2TN, UK
| | - Jacqueline Kim Dale
- Division of Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee DD1 4HN, UK
| | - Anestis Tsakiridis
- School of Biosciences, The University of Sheffield, Sheffield S10 2TN, UK
- Neuroscience Institute, The University of Sheffield, Sheffield S10 2TN, UK
| |
Collapse
|
7
|
Popova J, Bets V, Kozhevnikova E. Perspectives in Genome-Editing Techniques for Livestock. Animals (Basel) 2023; 13:2580. [PMID: 37627370 PMCID: PMC10452040 DOI: 10.3390/ani13162580] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/08/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Genome editing of farm animals has undeniable practical applications. It helps to improve production traits, enhances the economic value of livestock, and increases disease resistance. Gene-modified animals are also used for biomedical research and drug production and demonstrate the potential to be used as xenograft donors for humans. The recent discovery of site-specific nucleases that allow precision genome editing of a single-cell embryo (or embryonic stem cells) and the development of new embryological delivery manipulations have revolutionized the transgenesis field. These relatively new approaches have already proven to be efficient and reliable for genome engineering and have wide potential for use in agriculture. A number of advanced methodologies have been tested in laboratory models and might be considered for application in livestock animals. At the same time, these methods must meet the requirements of safety, efficiency and availability of their application for a wide range of farm animals. This review aims at covering a brief history of livestock animal genome engineering and outlines possible future directions to design optimal and cost-effective tools for transgenesis in farm species.
Collapse
Affiliation(s)
- Julia Popova
- Laboratory of Bioengineering, Novosibirsk State Agrarian University, 630039 Novosibirsk, Russia; (J.P.); (V.B.)
| | - Victoria Bets
- Laboratory of Bioengineering, Novosibirsk State Agrarian University, 630039 Novosibirsk, Russia; (J.P.); (V.B.)
- Center of Technological Excellence, Novosibirsk State Technical University, 630073 Novosibirsk, Russia
| | - Elena Kozhevnikova
- Laboratory of Bioengineering, Novosibirsk State Agrarian University, 630039 Novosibirsk, Russia; (J.P.); (V.B.)
- Laboratory of Experimental Models of Cognitive and Emotional Disorders, Scientific-Research Institute of Neurosciences and Medicine, 630117 Novosibirsk, Russia
| |
Collapse
|
8
|
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
|
9
|
Park JS, Woo SJ, Song CS, Han JY. Modification of surface glycan by expression of beta-1,4-N-acetyl-galactosaminyltransferase (B4GALNT2) confers resistance to multiple viruses infection in chicken fibroblast cell. Front Vet Sci 2023; 10:1160600. [PMID: 37483287 PMCID: PMC10358734 DOI: 10.3389/fvets.2023.1160600] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 06/19/2023] [Indexed: 07/25/2023] Open
Abstract
Introduction Infectious viruses in poultry, such as avian influenza virus (AIV) and Newcastle disease virus (NDV), are one of the most major threats to the poultry industry, resulting in enormous economic losses. AIVs and NDVs preferentially recognize α-2,3-linked sialic acid to bind to target cells. The human beta-1,4-N-acetyl-galactosaminyltransferase 2 (B4GALNT2) modifies α-2,3-linked sialic acid-containing glycan by transferring N-acetylgalactosamine to the sub-terminal galactose of the glycan, thus playing a pivotal role in preventing viruses from binding to cell surfaces. However, chickens lack a homolog of the B4GALNT2 gene. Methods Here, we precisely tagged the human B4GALNT2 gene downstream of the chicken GAPDH so that the engineered cells constitutively express the human B4GALNT2. We performed a lectin binding assay to analyze the modification of α-2,3-linked sialic acid-containing glycan by human B4GALNT2. Additionally, we infected the cells with AIV and NDV and compared cell survivability, viral gene transcription, and viral titer using the WST-1 assay, RT-qPCR and TCID50 assay, respectively. Results We validated human B4GALNT2 successfully modified α-2,3-linked sialic acid-containing glycan in chicken DF-1 cells. Following viral infection, we showed that human B4GALNT2 reduced infection of two AIV subtypes and NDV at 12-, 24-, and 36-hours post-infection. Moreover, cells expressing human B4GALNT2 showed significantly higher cell survivability compared to wild-type DF-1 cells, and viral gene expression was significantly reduced in the cells expressing human B4GALNT2. Discussion Collectively, these results suggest that artificially expressing human B4GALNT2 in chicken is a promising strategy to acquire broad resistance against infectious viruses with a preference for α-2,3-linked sialic acids such as AIV and NDV.
Collapse
Affiliation(s)
- Jin Se Park
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Seung Je Woo
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Chang Seon Song
- Avian Diseases Laboratory, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Jae Yong Han
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
10
|
Halasy V, Szőcs E, Soós Á, Kovács T, Pecsenye-Fejszák N, Hotta R, Goldstein AM, Nagy N. CXCR4 and CXCL12 signaling regulates the development of extrinsic innervation to the colorectum. Development 2023; 150:dev201289. [PMID: 37039233 PMCID: PMC10263150 DOI: 10.1242/dev.201289] [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: 09/09/2022] [Accepted: 01/25/2023] [Indexed: 04/12/2023]
Abstract
The gastrointestinal tract is innervated by an intrinsic neuronal network, known as the enteric nervous system (ENS), and by extrinsic axons arising from peripheral ganglia. The nerve of Remak (NoR) is an avian-specific sacral neural crest-derived ganglionated structure that extends from the cloaca to the proximal midgut and, similar to the pelvic plexus, provides extrinsic innervation to the distal intestine. The molecular mechanisms controlling extrinsic nerve fiber growth into the gut is unknown. In vertebrates, CXCR4, a cell-surface receptor for the CXCL12 chemokine, regulates migration of neural crest cells and axon pathfinding. We have employed chimeric tissue recombinations and organ culture assays to study the role of CXCR4 and CXCL12 molecules in the development of colorectal innervation. CXCR4 is specifically expressed in nerve fibers arising from the NoR and pelvic plexus, while CXCL12 is localized to the hindgut mesenchyme and enteric ganglia. Overexpression of CXCL12 results in significantly enhanced axonal projections to the gut from the NoR, while CXCR4 inhibition disrupts nerve fiber extension, supporting a previously unreported role for CXCR4 and CXCL12 signaling in extrinsic innervation of the colorectum.
Collapse
Affiliation(s)
- Viktória Halasy
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest 1094, Hungary
| | - Emőke Szőcs
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest 1094, Hungary
| | - Ádám Soós
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest 1094, Hungary
| | - Tamás Kovács
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest 1094, Hungary
| | - Nóra Pecsenye-Fejszák
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest 1094, Hungary
| | - Ryo Hotta
- Department of Pediatric Surgery, Pediatric Surgery Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Allan M. Goldstein
- Department of Pediatric Surgery, Pediatric Surgery Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Nándor Nagy
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest 1094, Hungary
| |
Collapse
|
11
|
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
|
12
|
Mizushima S, Sasanami T, Ono T, Kuroiwa A. Current Approaches to and the Application of Intracytoplasmic Sperm Injection (ICSI) for Avian Genome Editing. Genes (Basel) 2023; 14:genes14030757. [PMID: 36981028 PMCID: PMC10048369 DOI: 10.3390/genes14030757] [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: 02/23/2023] [Revised: 03/10/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Poultry are one of the most valuable resources for human society. They are also recognized as a powerful experimental animal for basic research on embryogenesis. Demands for the supply of low-allergen eggs and bioreactors have increased with the development of programmable genome editing technology. The CRISPR/Cas9 system has recently been used to produce transgenic animals and various animals in the agricultural industry and has also been successfully adopted for the modification of chicken and quail genomes. In this review, we describe the successful establishment of genome-edited lines combined with germline chimera production systems mediated by primordial germ cells and by viral infection in poultry. The avian intracytoplasmic sperm injection (ICSI) system that we previously established and recent advances in ICSI for genome editing are also summarized.
Collapse
Affiliation(s)
- Shusei Mizushima
- Faculty of Science, Hokkaido University, Kita 10 Nishi 8, Kita-ku, Sapporo 060-0810, Japan
| | - Tomohiro Sasanami
- Faculty of Agriculture, Shizuoka University, 836 Ohya, Shizuoka 422-8529, Japan
| | - Tamao Ono
- Matsumoto Dental University, 1780 Gobara, Hiro-oka, Shiojiri 399-0781, Nagano, Japan
| | - Asato Kuroiwa
- Faculty of Science, Hokkaido University, Kita 10 Nishi 8, Kita-ku, Sapporo 060-0810, Japan
| |
Collapse
|
13
|
Yahya I, Brand-Saberi B, Morosan-Puopolo G. Chicken embryo as a model in second heart field development. Heliyon 2023; 9:e14230. [PMID: 36923876 PMCID: PMC10009738 DOI: 10.1016/j.heliyon.2023.e14230] [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: 10/12/2022] [Revised: 01/30/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Previously, a single source of progenitor cells was thought to be responsible for the formation of the cardiac muscle. However, the second heart field has recently been identified as an additional source of myocardial progenitor cells. The chicken embryo, which develops in the egg, outside the mother can easily be manipulated in vivo and in vitro. Hence, it was an excellent model for establishing the concept of the second heart field. Here, our review will focus on the chicken model, specifically its role in understanding the second heart field. In addition to discussing historical aspects, we provide an overview of recent findings that have helped to define the chicken second heart field progenitor cells. A better understanding of the second heart field development will provide important insights into the congenital malformations affecting cardiac muscle formation and function.
Collapse
Affiliation(s)
- Imadeldin Yahya
- Department of Anatomy and Molecular Embryology, Ruhr University Bochum, 44801, Bochum, Germany
- Department of Anatomy, Faculty of Veterinary Medicine, University of Khartoum, Khartoum, 11115, Sudan
- Corresponding author. Department of Anatomy and Molecular Embryology, Ruhr University Bochum, 44801 Bochum, Germany.
| | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Ruhr University Bochum, 44801, Bochum, Germany
| | | |
Collapse
|
14
|
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
|
15
|
Abstract
Major advances in pathogen identification, treatment, vaccine development, and avian immunology have enabled the enormous expansion in global poultry production over the last 50 years. Looking forward, climate change, reduced feed, reduced water access, new avian pathogens and restrictions on the use of antimicrobials threaten to hamper further gains in poultry productivity and health. The development of novel in vitro cell culture systems, coupled with new genetic tools to investigate gene function, will aid in developing novel interventions for existing and newly emerging poultry pathogens. Our growing capacity to cryopreserve and generate genome-edited chicken lines will also be useful for developing improved chicken breeds for poultry farmers and conserving chicken genetic resources.
Collapse
Affiliation(s)
- Euan Mitchell
- The Roslin Institute & Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Guillermo Tellez
- The Roslin Institute & Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Mike J McGrew
- The Roslin Institute & Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
16
|
Smith J, Alfieri JM, Anthony N, Arensburger P, Athrey GN, Balacco J, Balic A, Bardou P, Barela P, Bigot Y, Blackmon H, Borodin PM, Carroll R, Casono MC, Charles M, Cheng H, Chiodi M, Cigan L, Coghill LM, Crooijmans R, Das N, Davey S, Davidian A, Degalez F, Dekkers JM, Derks M, Diack AB, Djikeng A, Drechsler Y, Dyomin A, Fedrigo O, Fiddaman SR, Formenti G, Frantz LA, Fulton JE, Gaginskaya E, Galkina S, Gallardo RA, Geibel J, Gheyas AA, Godinez CJP, Goodell A, Graves JA, Griffin DK, Haase B, Han JL, Hanotte O, Henderson LJ, Hou ZC, Howe K, Huynh L, Ilatsia E, Jarvis ED, Johnson SM, Kaufman J, Kelly T, Kemp S, Kern C, Keroack JH, Klopp C, Lagarrigue S, Lamont SJ, Lange M, Lanke A, Larkin DM, Larson G, Layos JKN, Lebrasseur O, Malinovskaya LP, Martin RJ, Martin Cerezo ML, Mason AS, McCarthy FM, McGrew MJ, Mountcastle J, Muhonja CK, Muir W, Muret K, Murphy TD, Ng'ang'a I, Nishibori M, O'Connor RE, Ogugo M, Okimoto R, Ouko O, Patel HR, Perini F, Pigozzi MI, Potter KC, Price PD, Reimer C, Rice ES, Rocos N, Rogers TF, Saelao P, Schauer J, Schnabel RD, Schneider VA, Simianer H, Smith A, et alSmith J, Alfieri JM, Anthony N, Arensburger P, Athrey GN, Balacco J, Balic A, Bardou P, Barela P, Bigot Y, Blackmon H, Borodin PM, Carroll R, Casono MC, Charles M, Cheng H, Chiodi M, Cigan L, Coghill LM, Crooijmans R, Das N, Davey S, Davidian A, Degalez F, Dekkers JM, Derks M, Diack AB, Djikeng A, Drechsler Y, Dyomin A, Fedrigo O, Fiddaman SR, Formenti G, Frantz LA, Fulton JE, Gaginskaya E, Galkina S, Gallardo RA, Geibel J, Gheyas AA, Godinez CJP, Goodell A, Graves JA, Griffin DK, Haase B, Han JL, Hanotte O, Henderson LJ, Hou ZC, Howe K, Huynh L, Ilatsia E, Jarvis ED, Johnson SM, Kaufman J, Kelly T, Kemp S, Kern C, Keroack JH, Klopp C, Lagarrigue S, Lamont SJ, Lange M, Lanke A, Larkin DM, Larson G, Layos JKN, Lebrasseur O, Malinovskaya LP, Martin RJ, Martin Cerezo ML, Mason AS, McCarthy FM, McGrew MJ, Mountcastle J, Muhonja CK, Muir W, Muret K, Murphy TD, Ng'ang'a I, Nishibori M, O'Connor RE, Ogugo M, Okimoto R, Ouko O, Patel HR, Perini F, Pigozzi MI, Potter KC, Price PD, Reimer C, Rice ES, Rocos N, Rogers TF, Saelao P, Schauer J, Schnabel RD, Schneider VA, Simianer H, Smith A, Stevens MP, Stiers K, Tiambo CK, Tixier-Boichard M, Torgasheva AA, Tracey A, Tregaskes CA, Vervelde L, Wang Y, Warren WC, Waters PD, Webb D, Weigend S, Wolc A, Wright AE, Wright D, Wu Z, Yamagata M, Yang C, Yin ZT, Young MC, Zhang G, Zhao B, Zhou H. Fourth Report on Chicken Genes and Chromosomes 2022. Cytogenet Genome Res 2023; 162:405-528. [PMID: 36716736 PMCID: PMC11835228 DOI: 10.1159/000529376] [Show More Authors] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 01/22/2023] [Indexed: 02/01/2023] Open
Affiliation(s)
- Jacqueline Smith
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Edinburgh, UK
| | - James M. Alfieri
- Interdisciplinary Program in Ecology and Evolutionary Biology, Texas A&M University, College Station, Texas, USA
- Department of Biology, Texas A&M University, College Station, Texas, USA
- Department of Poultry Science, Texas A&M University, College Station, Texas, USA
| | | | - Peter Arensburger
- Biological Sciences Department, California State Polytechnic University, Pomona, California, USA
| | - Giridhar N. Athrey
- Interdisciplinary Program in Ecology and Evolutionary Biology, Texas A&M University, College Station, Texas, USA
- Department of Poultry Science, Texas A&M University, College Station, Texas, USA
| | | | - Adam Balic
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Edinburgh, UK
| | - Philippe Bardou
- Université de Toulouse, INRAE, ENVT, GenPhySE, Sigenae, Castanet Tolosan, France
| | | | - Yves Bigot
- PRC, UMR INRAE 0085, CNRS 7247, Centre INRAE Val de Loire, Nouzilly, France
| | - Heath Blackmon
- Interdisciplinary Program in Ecology and Evolutionary Biology, Texas A&M University, College Station, Texas, USA
- Department of Biology, Texas A&M University, College Station, Texas, USA
| | - Pavel M. Borodin
- Department of Molecular Genetics, Cell Biology and Bioinformatics, Institute of Cytology and Genetics of Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russian Federation
| | - Rachel Carroll
- Department of Animal Sciences, Data Science and Informatics Institute, University of Missouri, Columbia, Missouri, USA
| | | | - Mathieu Charles
- University Paris-Saclay, INRAE, AgroParisTech, GABI, Sigenae, Jouy-en-Josas, France
| | - Hans Cheng
- USDA, ARS, USNPRC, Avian Disease and Oncology Laboratory, East Lansing, Michigan, USA
| | | | | | - Lyndon M. Coghill
- Department of Veterinary Pathology, University of Missouri, Columbia, Missouri, USA
| | - Richard Crooijmans
- Animal Breeding and Genomics, Wageningen University and Research, Wageningen, The Netherlands
| | | | - Sean Davey
- University of Arizona, Tucson, Arizona, USA
| | - Asya Davidian
- Saint Petersburg State University, Saint Petersburg, Russian Federation
| | - Fabien Degalez
- Centre for Tropical Livestock Genetics and Health (CTLGH) − ILRI, Nairobi, Kenya
| | - Jack M. Dekkers
- Department of Animal Science, University of California, Davis, California, USA
- INRAE, MIAT UR875, Sigenae, Castanet Tolosan, France
| | - Martijn Derks
- Animal Breeding and Genomics, Wageningen University and Research, Wageningen, The Netherlands
| | - Abigail B. Diack
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Edinburgh, UK
| | - Appolinaire Djikeng
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA
| | | | - Alexander Dyomin
- Saint Petersburg State University, Saint Petersburg, Russian Federation
| | | | | | | | - Laurent A.F. Frantz
- Queen Mary University of London, Bethnal Green, London, UK
- Palaeogenomics Group, Department of Veterinary Sciences, LMU Munich, Munich, Germany
| | - Janet E. Fulton
- Hy-Line International, Research and Development, Dallas Center, Iowa, USA
| | - Elena Gaginskaya
- Saint Petersburg State University, Saint Petersburg, Russian Federation
| | - Svetlana Galkina
- Saint Petersburg State University, Saint Petersburg, Russian Federation
| | - Rodrigo A. Gallardo
- School of Veterinary Medicine, University of California, Davis, California, USA
- Department of Animal Science, University of California, Davis, California, USA
| | - Johannes Geibel
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Neustadt, Germany
- Center for Integrated Breeding Research, University of Göttingen, Göttingen, Germany
| | - Almas A. Gheyas
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Edinburgh, UK
| | - Cyrill John P. Godinez
- Department of Animal Science, College of Agriculture and Food Science, Visayas State University, Baybay City, Philippines
| | | | - Jennifer A.M. Graves
- Department of Environment and Genetics, La Trobe University, Melbourne, Victoria, Australia
- Institute for Applied Ecology, University of Canberra, Canberra, Australian Capital Territory, Australia
| | | | | | - Jian-Lin Han
- CAAS-ILRI Joint Laboratory on Livestock and Forage Genetic Resources, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
- International Livestock Research Institute (ILRI), Addis Ababa, Ethiopia
| | - Olivier Hanotte
- International Livestock Research Institute (ILRI), Addis Ababa, Ethiopia
- Cells, Organisms and Molecular Genetics, School of Life Sciences, University of Nottingham, Nottingham, UK
- Centre for Tropical Livestock Genetics and Health, The Roslin Institute, Edinburgh, UK
| | - Lindsay J. Henderson
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Edinburgh, UK
| | - Zhuo-Cheng Hou
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, MARA, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | | | - Lan Huynh
- Institute for Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
| | - Evans Ilatsia
- Dairy Research Institute, Kenya Agricultural and Livestock Organization, Naivasha, Kenya
| | | | | | - Jim Kaufman
- Institute for Immunology and Infection Research, University of Edinburgh, Edinburgh, UK
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Terra Kelly
- School of Veterinary Medicine, University of California, Davis, California, USA
- Department of Animal Science, University of California, Davis, California, USA
| | - Steve Kemp
- INRAE, INSTITUT AGRO, PEGASE UMR 1348, Saint-Gilles, France
| | - Colin Kern
- Feed the Future Innovation Lab for Genomics to Improve Poultry, University of California, Davis, California, USA
| | | | - Christophe Klopp
- Department of Animal Science, Iowa State University, Ames, Iowa, USA
| | - Sandrine Lagarrigue
- Centre for Tropical Livestock Genetics and Health (CTLGH) − ILRI, Nairobi, Kenya
| | - Susan J. Lamont
- Department of Animal Science, University of California, Davis, California, USA
- INRAE, MIAT UR875, Sigenae, Castanet Tolosan, France
| | - Margaret Lange
- Centre for Tropical Livestock Genetics and Health (CTLGH) − The Roslin Institute, Edinburgh, UK
| | - Anika Lanke
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, California, USA
| | - Denis M. Larkin
- Department of Comparative Biomedical Sciences, Royal Veterinary College, University of London, London, UK
| | - Greger Larson
- The Palaeogenomics and Bio-Archaeology Research Network, Research Laboratory for Archaeology and History of Art, The University of Oxford, Oxford, UK
| | - John King N. Layos
- College of Agriculture and Forestry, Capiz State University, Mambusao, Philippines
| | - Ophélie Lebrasseur
- Centre d'Anthropobiologie et de Génomique de Toulouse (CAGT), CNRS UMR 5288, Université Toulouse III Paul Sabatier, Toulouse, France
- Instituto Nacional de Antropología y Pensamiento Latinoamericano, Ciudad Autónoma de Buenos Aires, Argentina
| | - Lyubov P. Malinovskaya
- Department of Cytology and Genetics, Novosibirsk State University, Novosibirsk, Russian Federation
| | - Rebecca J. Martin
- Saint Petersburg State University, Saint Petersburg, Russian Federation
| | | | | | | | - Michael J. McGrew
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Edinburgh, UK
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA
| | | | - Christine Kamidi Muhonja
- Department of Veterinary Pathology, University of Missouri, Columbia, Missouri, USA
- Centre for Tropical Livestock Genetics and Health (CTLGH) − ILRI, Nairobi, Kenya
| | - William Muir
- Department of Animal Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Kévin Muret
- Université Paris-Saclay, Commissariat à l'Energie Atomique et aux Energies Alternatives, Centre National de Recherche en Génomique Humaine, Evry, France
| | - Terence D. Murphy
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Masahide Nishibori
- Laboratory of Animal Genetics, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | | | - Moses Ogugo
- Centre for Tropical Livestock Genetics and Health (CTLGH) − ILRI, Nairobi, Kenya
| | - Ron Okimoto
- Cobb-Vantress, Siloam Springs, Arkansas, USA
| | - Ochieng Ouko
- Department of Veterinary Pathology, University of Missouri, Columbia, Missouri, USA
| | - Hardip R. Patel
- The John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Francesco Perini
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Edinburgh, UK
- Department of Agricultural, Food and Environmental Sciences, University of Perugia, Perugia, Italy
| | - María Ines Pigozzi
- INBIOMED (CONICET-UBA), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | - Peter D. Price
- Ecology and Evolutionary Biology, School of Biosciences, University of Sheffield, Sheffield, UK
| | - Christian Reimer
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Neustadt, Germany
| | - Edward S. Rice
- Department of Animal Sciences, Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA
| | - Nicolas Rocos
- USDA, ARS, USNPRC, Avian Disease and Oncology Laboratory, East Lansing, Michigan, USA
| | - Thea F. Rogers
- Department of Molecular Evolution and Development, University of Vienna, Vienna, Austria
| | - Perot Saelao
- Department of Animal Science, University of California, Davis, California, USA
- Veterinary Pest Genetics Research Unit, USDA, Kerrville, Texas, USA
| | - Jens Schauer
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Neustadt, Germany
| | - Robert D. Schnabel
- Department of Animal Sciences, University of Missouri, Columbia, Missouri, USA
| | - Valerie A. Schneider
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA
| | - Henner Simianer
- Center for Integrated Breeding Research, University of Göttingen, Göttingen, Germany
| | - Adrian Smith
- Department of Zoology, University of Oxford, Oxford, UK
| | - Mark P. Stevens
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Edinburgh, UK
| | - Kyle Stiers
- Department of Veterinary Pathology, University of Missouri, Columbia, Missouri, USA
| | | | | | - Anna A. Torgasheva
- Department of Molecular Genetics, Cell Biology and Bioinformatics, Institute of Cytology and Genetics of Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russian Federation
| | - Alan Tracey
- University Paris-Saclay, INRAE, AgroParisTech, GABI, Sigenae, Jouy-en-Josas, France
| | - Clive A. Tregaskes
- Animal Breeding and Genomics, Wageningen University and Research, Wageningen, The Netherlands
- Saint Petersburg State University, Saint Petersburg, Russian Federation
| | - Lonneke Vervelde
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Edinburgh, UK
| | - Ying Wang
- Department of Animal Science, University of California, Davis, California, USA
| | - Wesley C. Warren
- Department of Animal Sciences, Bond Life Sciences Center, University of Missouri, Columbia, Missouri, USA
- Department of Animal Sciences, University of Missouri, Columbia, Missouri, USA
| | - Paul D. Waters
- School of Biotechnology and Biomolecular Science, Faculty of Science, UNSW Sydney, Sydney, New South Wales, Australia
| | - David Webb
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA
| | - Steffen Weigend
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut, Neustadt, Germany
- Center for Integrated Breeding Research, University of Göttingen, Göttingen, Germany
| | - Anna Wolc
- INRAE, MIAT UR875, Sigenae, Castanet Tolosan, France
- Hy-Line International, Research and Development, Dallas Center, Iowa, USA
| | - Alison E. Wright
- Ecology and Evolutionary Biology, School of Biosciences, University of Sheffield, Sheffield, UK
| | - Dominic Wright
- AVIAN Behavioural Genomics and Physiology, IFM Biology, Linköping University, Linköping, Sweden
| | - Zhou Wu
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Edinburgh, UK
| | - Masahito Yamagata
- Center for Brain Science, Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts, USA
| | | | - Zhong-Tao Yin
- Department of Animal Sciences, Data Science and Informatics Institute, University of Missouri, Columbia, Missouri, USA
| | | | - Guojie Zhang
- Center for Evolutionary and Organismal Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Bingru Zhao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Huaijun Zhou
- Department of Animal Science, University of California, Davis, California, USA
| |
Collapse
|
17
|
Ibrahim M, Stadnicka K. The science of genetically modified poultry. PHYSICAL SCIENCES REVIEWS 2023. [DOI: 10.1515/psr-2022-0352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Abstract
The exuberant development of targeted genome editing has revolutionized research on the chicken genome, generating chickens with beneficial parameters. The chicken model is a crucial experimental tool that can be utilized for drug manufacture, preclinical research, pathological observation, and other applications. In essence, tweaking the chicken’s genome has enabled the poultry industry to get more done with less, generating genetically modified chickens that lay eggs containing large amounts of lifesaving humanized drugs. The transition of gene editing from concept to practical application has been dramatically hastened by the development of programmable nucleases, bringing scientists closer than ever to the efficient producers of tomorrow’s medicines. Combining the developmental and physiological characteristics of the chicken with cutting-edge genome editing, the chicken furnishes a potent frontier that is foreseen to be actively pursued in the future. Herein we review the current and future prospects of gene editing in chickens and the contributions to the development of humanized pharmaceuticals.
Collapse
Affiliation(s)
- Mariam Ibrahim
- Department of Animal Biotechnology and Genetics , PBS University of Science and Technology , 85-084 Bydgoszcz , Poland
| | - Katarzyna Stadnicka
- Department of Oncology , Collegium Medicum Nicolaus Copernicus University , 85-821 Bydgoszcz , Poland
| |
Collapse
|
18
|
Berki B, Sacher F, Fages A, Tschopp P, Luxey M. A method to investigate muscle target-specific transcriptional signatures of single motor neurons. Dev Dyn 2023; 252:208-219. [PMID: 35705847 PMCID: PMC10084336 DOI: 10.1002/dvdy.507] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Motor neurons in the vertebrate spinal cord have long served as a paradigm to study the transcriptional logic of cell type specification and differentiation. At limb levels, pool-specific transcriptional signatures first restrict innervation to only one particular muscle in the periphery, and get refined, once muscle connection has been established. Accordingly, to study the transcriptional dynamics and specificity of the system, a method for establishing muscle target-specific motor neuron transcriptomes would be required. RESULTS To investigate target-specific transcriptional signatures of single motor neurons, here we combine ex-ovo retrograde axonal labeling in mid-gestation chicken embryos with manual isolation of individual fluorescent cells and Smart-seq2 single-cell RNA-sequencing. We validate our method by injecting the dorsal extensor metacarpi radialis and ventral flexor digiti quarti wing muscles and harvesting a total of 50 fluorescently labeled cells, in which we detect up to 12,000 transcribed genes. Additionally, we present visual cues and cDNA metrics predictive of sequencing success. CONCLUSIONS Our method provides a unique approach to study muscle target-specific motor neuron transcriptomes at a single-cell resolution. We anticipate that our method will provide key insights into the transcriptional logic underlying motor neuron pool specialization and proper neuromuscular circuit assembly and refinement.
Collapse
Affiliation(s)
- Bianka Berki
- DUW Zoology, University of Basel, Basel, Switzerland
| | - Fabio Sacher
- DUW Zoology, University of Basel, Basel, Switzerland
| | - Antoine Fages
- DUW Zoology, University of Basel, Basel, Switzerland
| | | | - Maëva Luxey
- DUW Zoology, University of Basel, Basel, Switzerland
| |
Collapse
|
19
|
Gul H, Habib G, Khan IM, Rahman SU, Khan NM, Wang H, Khan NU, Liu Y. Genetic resilience in chickens against bacterial, viral and protozoal pathogens. Front Vet Sci 2022; 9:1032983. [PMID: 36439341 PMCID: PMC9691405 DOI: 10.3389/fvets.2022.1032983] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/28/2022] [Indexed: 06/13/2024] Open
Abstract
The genome contributes to the uniqueness of an individual breed, and enables distinctive characteristics to be passed from one generation to the next. The allelic heterogeneity of a certain breed results in a different response to a pathogen with different genomic expression. Disease resistance in chicken is a polygenic trait that involves different genes that confer resistance against pathogens. Such resistance also involves major histocompatibility (MHC) molecules, immunoglobulins, cytokines, interleukins, T and B cells, and CD4+ and CD8+ T lymphocytes, which are involved in host protection. The MHC is associated with antigen presentation, antibody production, and cytokine stimulation, which highlight its role in disease resistance. The natural resistance-associated macrophage protein 1 (Nramp-1), interferon (IFN), myxovirus-resistance gene, myeloid differentiation primary response 88 (MyD88), receptor-interacting serine/threonine kinase 2 (RIP2), and heterophile cells are involved in disease resistance and susceptibility of chicken. Studies related to disease resistance genetics, epigenetics, and quantitative trait loci would enable the identification of resistance markers and the development of disease resistance breeds. Microbial infections are responsible for significant outbreaks and have blighted the poultry industry. Breeding disease-resistant chicken strains may be helpful in tackling pathogens and increasing the current understanding on host genetics in the fight against communicable diseases. Advanced technologies, such as the CRISPR/Cas9 system, whole genome sequencing, RNA sequencing, and high-density single nucleotide polymorphism (SNP) genotyping, aid the development of resistant breeds, which would significantly decrease the use of antibiotics and vaccination in poultry. In this review, we aimed to reveal the recent genetic basis of infection and genomic modification that increase resistance against different pathogens in chickens.
Collapse
Affiliation(s)
- Haji Gul
- Anhui Province Key Laboratory of Embryo Development and Reproduction Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, School of Biological and Food Engineering, Fuyang Normal University, Fuyang, China
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Gul Habib
- Department of Microbiology, Abbottabad University of Science and Technology, Abbottabad, Pakistan
| | - Ibrar Muhammad Khan
- Anhui Province Key Laboratory of Embryo Development and Reproduction Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, School of Biological and Food Engineering, Fuyang Normal University, Fuyang, China
| | - Sajid Ur Rahman
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Animal Parasitology of Ministry of Agriculture, Laboratory of Quality and Safety Risk Assessment for Animal Products on Biohazards (Shanghai) of Ministry of Agriculture, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Nazir Muhammad Khan
- Department of Zoology, University of Science and Technology, Bannu, Pakistan
| | - Hongcheng Wang
- Anhui Province Key Laboratory of Embryo Development and Reproduction Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, School of Biological and Food Engineering, Fuyang Normal University, Fuyang, China
| | - Najeeb Ullah Khan
- Institute of Biotechnology and Genetic Engineering, The University of Agriculture, Peshawar, Pakistan
| | - Yong Liu
- Anhui Province Key Laboratory of Embryo Development and Reproduction Regulation, Anhui Province Key Laboratory of Environmental Hormone and Reproduction, School of Biological and Food Engineering, Fuyang Normal University, Fuyang, China
| |
Collapse
|
20
|
Panda SK, McGrew MJ. Genome editing of avian species: implications for animal use and welfare. Lab Anim 2022; 56:50-59. [PMID: 33691522 PMCID: PMC8918865 DOI: 10.1177/0023677221998400] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 01/05/2023]
Abstract
Avian species are used as model systems in research and have contributed to ground-breaking concepts in developmental biology, immunology, genetics, virology, cancer and cell biology. The chicken in particular is an important research model and an agricultural animal as a major contributor to animal protein resources for the global population. The development of genome editing methods, including CRISPR/Cas9, to mediate germline engineering of the avian genome will have important applications in biomedical, agricultural and biotechnological activities. Notably, these precise genome editing tools have the potential to enhance avian health and productivity by identifying and validating beneficial genetic variants in bird populations. Here, we present a concise description of the existing methods and current applications of the genome editing tools in bird species, focused on chickens, with attention on animal use and welfare issues for each of the techniques presented.
Collapse
Affiliation(s)
- Sudeepta K Panda
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, UK
| | - Mike J McGrew
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, UK
| |
Collapse
|
21
|
Abstract
For four decades, genetically altered laboratory animals have provided invaluable information. Originally, genetic modifications were performed on only a few animal species, often chosen because of the ready accessibility of embryonic materials and short generation times. The methods were often slow, inefficient and expensive. In 2013, a new, extremely efficient technology, namely CRISPR/Cas9, not only made the production of genetically altered organisms faster and cheaper, but also opened it up to non-conventional laboratory animal species. CRISPR/Cas9 relies on a guide RNA as a 'location finder' to target DNA double strand breaks induced by the Cas9 enzyme. This is a prerequisite for non-homologous end joining repair to occur, an error prone mechanism often generating insertion or deletion of genetic material. If a DNA template is also provided, this can lead to homology directed repair, allowing precise insertions, deletions or substitutions. Due to its high efficiency in targeting DNA, CRISPR/Cas9-mediated genetic modification is now possible in virtually all animal species for which we have genome sequence data. Furthermore, modifications of Cas9 have led to more refined genetic alterations from targeted single base-pair mutations to epigenetic modifications. The latter offer altered gene expression without genome alteration. With this ever growing genetic toolbox, the number and range of genetically altered conventional and non-conventional laboratory animals with simple or complex genetic modifications is growing exponentially.
Collapse
|
22
|
Delalande JM, Nagy N, McCann CJ, Natarajan D, Cooper JE, Carreno G, Dora D, Campbell A, Laurent N, Kemos P, Thomas S, Alby C, Attié-Bitach T, Lyonnet S, Logan MP, Goldstein AM, Davey MG, Hofstra RMW, Thapar N, Burns AJ. TALPID3/KIAA0586 Regulates Multiple Aspects of Neuromuscular Patterning During Gastrointestinal Development in Animal Models and Human. Front Mol Neurosci 2022; 14:757646. [PMID: 35002618 PMCID: PMC8733242 DOI: 10.3389/fnmol.2021.757646] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 11/10/2021] [Indexed: 12/26/2022] Open
Abstract
TALPID3/KIAA0586 is an evolutionary conserved protein, which plays an essential role in protein trafficking. Its role during gastrointestinal (GI) and enteric nervous system (ENS) development has not been studied previously. Here, we analyzed chicken, mouse and human embryonic GI tissues with TALPID3 mutations. The GI tract of TALPID3 chicken embryos was shortened and malformed. Histologically, the gut smooth muscle was mispatterned and enteric neural crest cells were scattered throughout the gut wall. Analysis of the Hedgehog pathway and gut extracellular matrix provided causative reasons for these defects. Interestingly, chicken intra-species grafting experiments and a conditional knockout mouse model showed that ENS formation did not require TALPID3, but was dependent on correct environmental cues. Surprisingly, the lack of TALPID3 in enteric neural crest cells (ENCC) affected smooth muscle and epithelial development in a non-cell-autonomous manner. Analysis of human gut fetal tissues with a KIAA0586 mutation showed strikingly similar findings compared to the animal models demonstrating conservation of TALPID3 and its necessary role in human GI tract development and patterning.
Collapse
Affiliation(s)
- Jean Marie Delalande
- Centre for Immunobiology, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.,Stem Cells and Regenerative Medicine, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Nandor Nagy
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Conor J McCann
- Stem Cells and Regenerative Medicine, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Dipa Natarajan
- Stem Cells and Regenerative Medicine, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Julie E Cooper
- Developmental Biology and Cancer Program, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Gabriela Carreno
- Developmental Biology and Cancer Program, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - David Dora
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Alison Campbell
- Department of Paediatric Surgery, Christchurch Hospital, Christchurch, New Zealand
| | - Nicole Laurent
- Génétique et Anomalies du Développement, Université de Bourgogne, Service d'Anatomie Pathologique, Dijon, France
| | - Polychronis Kemos
- Centre for Immunobiology, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Sophie Thomas
- Laboratory of Embryology and Genetics of Congenital Malformations, INSERM UMR 1163 Institut Imagine, Paris, France
| | - Caroline Alby
- Department of Genetics, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris (AP-HP), Paris, France
| | - Tania Attié-Bitach
- Laboratory of Embryology and Genetics of Congenital Malformations, INSERM UMR 1163 Institut Imagine, Paris, France.,Department of Genetics, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris (AP-HP), Paris, France.,Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Stanislas Lyonnet
- Laboratory of Embryology and Genetics of Congenital Malformations, INSERM UMR 1163 Institut Imagine, Paris, France.,Department of Genetics, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris (AP-HP), Paris, France.,Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Malcolm P Logan
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Allan M Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Megan G Davey
- Division of Developmental Biology, The Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Robert M W Hofstra
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Nikhil Thapar
- Stem Cells and Regenerative Medicine, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom.,Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Alan J Burns
- Stem Cells and Regenerative Medicine, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom.,Division of Neurogastroenterology and Motility, Department of Gastroenterology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom.,Gastrointestinal Drug Discovery Unit, Takeda Pharmaceuticals International, Inc., Cambridge, MA, United States
| |
Collapse
|
23
|
Nagy N, Kovacs T, Stavely R, Halasy V, Soos A, Szocs E, Hotta R, Graham H, Goldstein AM. Avian ceca are indispensable for hindgut enteric nervous system development. Development 2021; 148:dev199825. [PMID: 34792104 PMCID: PMC8645208 DOI: 10.1242/dev.199825] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 10/22/2021] [Indexed: 11/20/2022]
Abstract
The enteric nervous system (ENS), which is derived from enteric neural crest cells (ENCCs), represents the neuronal innervation of the intestine. Compromised ENCC migration can lead to Hirschsprung disease, which is characterized by an aganglionic distal bowel. During the craniocaudal migration of ENCCs along the gut, we find that their proliferation is greatest as the ENCC wavefront passes through the ceca, a pair of pouches at the midgut-hindgut junction in avian intestine. Removal of the ceca leads to hindgut aganglionosis, suggesting that they are required for ENS development. Comparative transcriptome profiling of the cecal buds compared with the interceca region shows that the non-canonical Wnt signaling pathway is preferentially expressed within the ceca. Specifically, WNT11 is highly expressed, as confirmed by RNA in situ hybridization, leading us to hypothesize that cecal expression of WNT11 is important for ENCC colonization of the hindgut. Organ cultures using embryonic day 6 avian intestine show that WNT11 inhibits enteric neuronal differentiation. These results reveal an essential role for the ceca during hindgut ENS formation and highlight an important function for non-canonical Wnt signaling in regulating ENCC differentiation.
Collapse
Affiliation(s)
- Nandor Nagy
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, 1094, Hungary
| | - Tamas Kovacs
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, 1094, Hungary
| | - Rhian Stavely
- Department of Pediatric Surgery, Pediatric Surgery Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114,USA
| | - Viktoria Halasy
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, 1094, Hungary
| | - Adam Soos
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, 1094, Hungary
| | - Emoke Szocs
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, 1094, Hungary
| | - Ryo Hotta
- Department of Pediatric Surgery, Pediatric Surgery Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114,USA
| | - Hannah Graham
- Department of Pediatric Surgery, Pediatric Surgery Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114,USA
| | - Allan M. Goldstein
- Department of Pediatric Surgery, Pediatric Surgery Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114,USA
| |
Collapse
|
24
|
Flores-Santin J, Burggren WW. Beyond the Chicken: Alternative Avian Models for Developmental Physiological Research. Front Physiol 2021; 12:712633. [PMID: 34744759 PMCID: PMC8566884 DOI: 10.3389/fphys.2021.712633] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 09/13/2021] [Indexed: 12/23/2022] Open
Abstract
Biomedical research focusing on physiological, morphological, behavioral, and other aspects of development has long depended upon the chicken (Gallus gallus domesticus) as a key animal model that is presumed to be typical of birds and generally applicable to mammals. Yet, the modern chicken in its many forms is the result of artificial selection more intense than almost any other domesticated animal. A consequence of great variation in genotype and phenotype is that some breeds have inherent aberrant physiological and morphological traits that may show up relatively early in development (e.g., hypertension, hyperglycemia, and limb defects in the broiler chickens). While such traits can be useful as models of specific diseases, this high degree of specialization can color general experimental results and affect their translational value. Against this background, in this review we first consider the characteristics that make an animal model attractive for developmental research (e.g., accessibility, ease of rearing, size, fecundity, development rates, genetic variation, etc.). We then explore opportunities presented by the embryo to adult continuum of alternative bird models, including quail, ratites, songbirds, birds of prey, and corvids. We conclude by indicating that expanding developmental studies beyond the chicken model to include additional avian groups will both validate the chicken model as well as potentially identify even more suitable avian models for answering questions applicable to both basic biology and the human condition.
Collapse
Affiliation(s)
- Josele Flores-Santin
- Facultad de Ciencias, Biologia, Universidad Autónoma del Estado de Mexico, Toluca, Mexico
| | - Warren W. Burggren
- Developmental Integrative Biology Research Group, Department of Biological Sciences, University of North Texas Denton, Denton, TX, United States
| |
Collapse
|
25
|
Dehdilani N, Taemeh SY, Goshayeshi L, Dehghani H. Genetically engineered birds; pre-CRISPR and CRISPR era. Biol Reprod 2021; 106:24-46. [PMID: 34668968 DOI: 10.1093/biolre/ioab196] [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: 08/12/2021] [Revised: 10/08/2021] [Accepted: 10/14/2021] [Indexed: 11/14/2022] Open
Abstract
Generating biopharmaceuticals in genetically engineered bioreactors continues to reign supreme. Hence, genetically engineered birds have attracted considerable attention from the biopharmaceutical industry. Fairly recent genome engineering methods have made genome manipulation an easy and affordable task. In this review, we first provide a broad overview of the approaches and main impediments ahead of generating efficient and reliable genetically engineered birds, and various factors that affect the fate of a transgene. This section provides an essential background for the rest of the review, in which we discuss and compare different genome manipulation methods in the pre-CRISPR and CRISPR era in the field of avian genome engineering.
Collapse
Affiliation(s)
- Nima Dehdilani
- Stem Cell Biology and Regenerative Medicine Research Group, Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Sara Yousefi Taemeh
- Stem Cell Biology and Regenerative Medicine Research Group, Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Lena Goshayeshi
- Stem Cell Biology and Regenerative Medicine Research Group, Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hesam Dehghani
- Stem Cell Biology and Regenerative Medicine Research Group, Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.,Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran.,Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
26
|
Estermann MA, Major AT, Smith CA. Genetic Regulation of Avian Testis Development. Genes (Basel) 2021; 12:1459. [PMID: 34573441 PMCID: PMC8470383 DOI: 10.3390/genes12091459] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/16/2021] [Accepted: 09/16/2021] [Indexed: 11/30/2022] Open
Abstract
As in other vertebrates, avian testes are the site of spermatogenesis and androgen production. The paired testes of birds differentiate during embryogenesis, first marked by the development of pre-Sertoli cells in the gonadal primordium and their condensation into seminiferous cords. Germ cells become enclosed in these cords and enter mitotic arrest, while steroidogenic Leydig cells subsequently differentiate around the cords. This review describes our current understanding of avian testis development at the cell biology and genetic levels. Most of this knowledge has come from studies on the chicken embryo, though other species are increasingly being examined. In chicken, testis development is governed by the Z-chromosome-linked DMRT1 gene, which directly or indirectly activates the male factors, HEMGN, SOX9 and AMH. Recent single cell RNA-seq has defined cell lineage specification during chicken testis development, while comparative studies point to deep conservation of avian testis formation. Lastly, we identify areas of future research on the genetics of avian testis development.
Collapse
Affiliation(s)
| | | | - Craig Allen Smith
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; (M.A.E.); (A.T.M.)
| |
Collapse
|
27
|
Karachaliou CE, Vassilakopoulou V, Livaniou E. IgY technology: Methods for developing and evaluating avian immunoglobulins for the in vitro detection of biomolecules. World J Methodol 2021; 11:243-262. [PMID: 34631482 PMCID: PMC8472547 DOI: 10.5662/wjm.v11.i5.243] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 06/10/2021] [Accepted: 07/13/2021] [Indexed: 02/06/2023] Open
Abstract
The term “IgY technology” was introduced in the literature in the mid 1990s to describe a procedure involving immunization of avian species, mainly laying hens and consequent isolation of the polyclonal IgYs from the “immune” egg yolk (thus avoiding bleeding and animal stress). IgYs have been applied to various fields of medicine and biotechnology. The present article will deal with specific aspects of IgY technology, focusing on the currently reported methods for developing, isolating, evaluating and storing polyclonal IgYs. Other topics such as current information on isolation protocols or evaluation of IgYs from different avian species are also discussed. Specific advantages of IgY technology (e.g., novel antibody specificities that may emerge via the avian immune system) will also be discussed. Recent in vitro applications of polyclonal egg yolk-derived IgYs to the field of disease diagnosis in human and veterinary medicine through in vitro immunodetection of target biomolecules will be presented. Moreover, ethical aspects associated with animal well-being as well as new promising approaches that are relevant to the original IgY technology (e.g., development of monoclonal IgYs and IgY-like antibodies through the phage display technique or in transgenic chickens) and future prospects in the area will also be mentioned.
Collapse
Affiliation(s)
- Chrysoula-Evangelia Karachaliou
- Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Centre for Scientific Research “Demokritos”, Athens 15310, Greece
| | - Vyronia Vassilakopoulou
- Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Centre for Scientific Research “Demokritos”, Athens 15310, Greece
| | - Evangelia Livaniou
- Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Centre for Scientific Research “Demokritos”, Athens 15310, Greece
| |
Collapse
|
28
|
Guillot C, Djeffal Y, Michaut A, Rabe B, Pourquié O. Dynamics of primitive streak regression controls the fate of neuromesodermal progenitors in the chicken embryo. eLife 2021; 10:64819. [PMID: 34227938 PMCID: PMC8260230 DOI: 10.7554/elife.64819] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 06/23/2021] [Indexed: 12/20/2022] Open
Abstract
In classical descriptions of vertebrate development, the segregation of the three embryonic germ layers completes by the end of gastrulation. Body formation then proceeds in a head to tail fashion by progressive deposition of lineage-committed progenitors during regression of the primitive streak (PS) and tail bud (TB). The identification by retrospective clonal analysis of a population of neuromesodermal progenitors (NMPs) contributing to both musculoskeletal precursors (paraxial mesoderm) and spinal cord during axis formation challenged these notions. However, classical fate mapping studies of the PS region in amniotes have so far failed to provide direct evidence for such bipotential cells at the single-cell level. Here, using lineage tracing and single-cell RNA sequencing in the chicken embryo, we identify a resident cell population of the anterior PS epiblast, which contributes to neural and mesodermal lineages in trunk and tail. These cells initially behave as monopotent progenitors as classically described and only acquire a bipotential fate later, in more posterior regions. We show that NMPs exhibit a conserved transcriptomic signature during axis elongation but lose their epithelial characteristicsin the TB. Posterior to anterior gradients of convergence speed and ingression along the PS lead to asymmetric exhaustion of PS mesodermal precursor territories. Through limited ingression and increased proliferation, NMPs are maintained and amplified as a cell population which constitute the main progenitors in the TB. Together, our studies provide a novel understanding of the PS and TB contribution through the NMPs to the formation of the body of amniote embryos.
Collapse
Affiliation(s)
- Charlene Guillot
- Department of Pathology, Brigham and Women's Hospital, Boston, United States.,Department of Genetics, Harvard Medical School, Boston, United States.,Harvard Stem Cell Institute, Boston, United States
| | - Yannis Djeffal
- Department of Pathology, Brigham and Women's Hospital, Boston, United States.,Department of Genetics, Harvard Medical School, Boston, United States.,Harvard Stem Cell Institute, Boston, United States
| | - Arthur Michaut
- Department of Pathology, Brigham and Women's Hospital, Boston, United States.,Department of Genetics, Harvard Medical School, Boston, United States.,Harvard Stem Cell Institute, Boston, United States
| | - Brian Rabe
- Department of Genetics, Harvard Medical School, Boston, United States.,Howard Hughes Medical Institute, Boston, United States
| | - Olivier Pourquié
- Department of Pathology, Brigham and Women's Hospital, Boston, United States.,Department of Genetics, Harvard Medical School, Boston, United States.,Harvard Stem Cell Institute, Boston, United States
| |
Collapse
|
29
|
Yang H, Lee BR, Lee HC, Choi H, Jung SK, Kim JY, No J, Shanmugam S, Jo YJ, Oh KB, Kim KW, Byun SJ. Development and in vitro evaluation of recombinant chicken promoters to efficiently drive transgene expression in chicken oviduct cells. Poult Sci 2021; 100:101365. [PMID: 34375836 PMCID: PMC8358702 DOI: 10.1016/j.psj.2021.101365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 06/19/2021] [Accepted: 06/24/2021] [Indexed: 12/04/2022] Open
Abstract
Virus injection into EGK-X embryos is a well-defined approach in avian transgenesis. This system uses a chicken ovalbumin gene promoter to induce transgene expression in the chicken oviduct. Although a reconstructed chicken ovalbumin promoter that links an ovalbumin promoter and estrogen-responsive enhancer element (ERE) is useful, a large viral vector containing the ovalbumin promoter and a target gene restricts viral packaging capacity and produces low-titer virus particles. We newly developed recombinant chicken promoters by linking regulatory regions of ovalbumin and other oviduct-specific genes. Putative enhancer fragments of the genes, such as ovotransferrin (TF), ovomucin alpha subunit (OVOA), and ovalbumin-related protein X (OVALX), were placed at the 5`-flanking region of the 2.8-kb ovalbumin promoter. Basal promoter fragments of the genes, namely, pTF, lysozyme (pLYZ), and ovomucoid (pOVM), were placed at the 3`-flanking region of the 1.6-kb ovalbumin ERE. The recombinant promoters cloned into each reporter vector were evaluated using a dual luciferase assay in human and chicken somatic cells, and LMH/2A cells treated with 0-1,000 nM estrogen, and cultured primary chicken oviduct cells. The recombinant promoters with linking ovalbumin and TF, OVOA, pOVM, and pLYZ regulatory regions had 2.1- to 19.5-fold (P < 0.05) higher luciferase activity than the reconstructed ovalbumin promoter in chicken oviduct cells. Therefore, recombinant promoters may be used to efficiently drive transgene expression in transgenic chickens.
Collapse
Affiliation(s)
- Hyeon Yang
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun 55365, Republic of Korea
| | - Bo Ram Lee
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun 55365, Republic of Korea
| | - Hwi-Cheul Lee
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun 55365, Republic of Korea
| | - Hoonsung Choi
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun 55365, Republic of Korea
| | - Sun Keun Jung
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun 55365, Republic of Korea
| | - Ji-Youn Kim
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun 55365, Republic of Korea
| | - Jingu No
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun 55365, Republic of Korea
| | - Sureshkumar Shanmugam
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun 55365, Republic of Korea
| | - Yong Jin Jo
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun 55365, Republic of Korea
| | - Keon Bong Oh
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun 55365, Republic of Korea
| | - Kyung Woon Kim
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun 55365, Republic of Korea
| | - Sung June Byun
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun 55365, Republic of Korea.
| |
Collapse
|
30
|
Esteves de Lima J, Blavet C, Bonnin MA, Hirsinger E, Comai G, Yvernogeau L, Delfini MC, Bellenger L, Mella S, Nassari S, Robin C, Schweitzer R, Fournier-Thibault C, Jaffredo T, Tajbakhsh S, Relaix F, Duprez D. Unexpected contribution of fibroblasts to muscle lineage as a mechanism for limb muscle patterning. Nat Commun 2021; 12:3851. [PMID: 34158501 PMCID: PMC8219714 DOI: 10.1038/s41467-021-24157-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 06/06/2021] [Indexed: 12/13/2022] Open
Abstract
Positional information driving limb muscle patterning is contained in connective tissue fibroblasts but not in myogenic cells. Limb muscles originate from somites, while connective tissues originate from lateral plate mesoderm. With cell and genetic lineage tracing we challenge this model and identify an unexpected contribution of lateral plate-derived fibroblasts to the myogenic lineage, preferentially at the myotendinous junction. Analysis of single-cell RNA-sequencing data from whole limbs at successive developmental stages identifies a population displaying a dual muscle and connective tissue signature. BMP signalling is active in this dual population and at the tendon/muscle interface. In vivo and in vitro gain- and loss-of-function experiments show that BMP signalling regulates a fibroblast-to-myoblast conversion. These results suggest a scenario in which BMP signalling converts a subset of lateral plate mesoderm-derived cells to a myogenic fate in order to create a boundary of fibroblast-derived myonuclei at the myotendinous junction that controls limb muscle patterning.
Collapse
Affiliation(s)
- Joana Esteves de Lima
- Developmental Biology Laboratory, Institut Biologie Paris Seine, Sorbonne Université, CNRS, IBPS-UMR 7622, Paris, France
- Inserm U1156, Paris, France
- Univ Paris Est Creteil, INSERM, EnvA, EFS, AP-HP, IMRB, Creteil, France
| | - Cédrine Blavet
- Developmental Biology Laboratory, Institut Biologie Paris Seine, Sorbonne Université, CNRS, IBPS-UMR 7622, Paris, France
- Inserm U1156, Paris, France
| | - Marie-Ange Bonnin
- Developmental Biology Laboratory, Institut Biologie Paris Seine, Sorbonne Université, CNRS, IBPS-UMR 7622, Paris, France
- Inserm U1156, Paris, France
| | - Estelle Hirsinger
- Developmental Biology Laboratory, Institut Biologie Paris Seine, Sorbonne Université, CNRS, IBPS-UMR 7622, Paris, France
- Inserm U1156, Paris, France
| | - Glenda Comai
- Department of Developmental and Stem Cell Biology, Institut Pasteur, CNRS UMR 3738, Paris, France
| | - Laurent Yvernogeau
- Developmental Biology Laboratory, Institut Biologie Paris Seine, Sorbonne Université, CNRS, IBPS-UMR 7622, Paris, France
- Inserm U1156, Paris, France
- Hubrecht Institute-Royal Netherlands Academy of Arts and Sciences (KNAW), Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marie-Claire Delfini
- Developmental Biology Laboratory, Institut Biologie Paris Seine, Sorbonne Université, CNRS, IBPS-UMR 7622, Paris, France
- Inserm U1156, Paris, France
- Aix Marseille University, CNRS, IBDM, Marseille, France
| | - Léa Bellenger
- Institut Biologie Paris Seine, Sorbonne Université, CNRS, IBPS-FR3631, ARTbio Bioinformatics Platform, Inserm US 037, Paris, France
| | - Sébastien Mella
- Department of Developmental and Stem Cell Biology, Institut Pasteur, CNRS UMR 3738, Paris, France
| | - Sonya Nassari
- Developmental Biology Laboratory, Institut Biologie Paris Seine, Sorbonne Université, CNRS, IBPS-UMR 7622, Paris, France
- Inserm U1156, Paris, France
| | - Catherine Robin
- Hubrecht Institute-Royal Netherlands Academy of Arts and Sciences (KNAW), Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ronen Schweitzer
- Research Division, Shriners Hospital for Children, Portland, OR, USA
| | - Claire Fournier-Thibault
- Developmental Biology Laboratory, Institut Biologie Paris Seine, Sorbonne Université, CNRS, IBPS-UMR 7622, Paris, France
- Inserm U1156, Paris, France
| | - Thierry Jaffredo
- Developmental Biology Laboratory, Institut Biologie Paris Seine, Sorbonne Université, CNRS, IBPS-UMR 7622, Paris, France
- Inserm U1156, Paris, France
| | - Shahragim Tajbakhsh
- Department of Developmental and Stem Cell Biology, Institut Pasteur, CNRS UMR 3738, Paris, France
| | - Frédéric Relaix
- Univ Paris Est Creteil, INSERM, EnvA, EFS, AP-HP, IMRB, Creteil, France
| | - Delphine Duprez
- Developmental Biology Laboratory, Institut Biologie Paris Seine, Sorbonne Université, CNRS, IBPS-UMR 7622, Paris, France.
- Inserm U1156, Paris, France.
| |
Collapse
|
31
|
Why zebra finches don't get hypercholesterolemia. Proc Natl Acad Sci U S A 2021; 118:2107021118. [PMID: 34011675 DOI: 10.1073/pnas.2107021118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
32
|
Improving germline transmission efficiency in chimeric chickens using a multi-stage injection approach. PLoS One 2021; 16:e0247471. [PMID: 34086696 PMCID: PMC8177527 DOI: 10.1371/journal.pone.0247471] [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: 02/07/2021] [Accepted: 05/18/2021] [Indexed: 11/19/2022] Open
Abstract
Although different strategies have been developed to generate transgenic poultry, low efficiency of germline transgene transmission has remained a challenge in poultry transgenesis. Herein, we developed an efficient germline transgenesis method using a lentiviral vector system in chickens through multiple injections of transgenes into embryos at different stages of development. The embryo chorioallantoic membrane (CAM) vasculature was successfully used as a novel route of gene transfer into germline tissues. Compared to the other routes of viral vector administration, the embryo’s bloodstream at Hamburger-Hamilton (HH) stages 14–15 achieved the highest rate of germline transmission (GT), 7.7%. Single injection of viral vectors into the CAM vasculature resulted in a GT efficiency of 2.7%, which was significantly higher than the 0.4% obtained by injection into embryos at the blastoderm stage. Double injection of viral vectors into the bloodstream at HH stages 14–15 and through CAM was the most efficient method for producing germline chimeras, giving a GT rate of 13.6%. The authors suggest that the new method described in this study could be efficiently used to produce transgenic poultry in virus-mediated gene transfer systems.
Collapse
|
33
|
Divergent low-density lipoprotein receptor (LDLR) linked to low VSV G-dependent viral infectivity and unique serum lipid profile in zebra finches. Proc Natl Acad Sci U S A 2021; 118:2025167118. [PMID: 33903244 PMCID: PMC8106303 DOI: 10.1073/pnas.2025167118] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The low-density lipoprotein receptor (LDLR) is key to cellular cholesterol uptake and is also the main receptor for the vesicular stomatitis virus glycoprotein (VSV G). Here we show that in songbirds LDLR is highly divergent and lacks domains critical for ligand binding and cellular trafficking, inconsistent with universal structure conservation and function across vertebrates. Linked to the LDLR functional domain loss, zebra finches show inefficient infectivity by lentiviruses (LVs) pseudotyped with VSV G, which can be rescued by the expression of human LDLR. Finches also show an atypical plasma lipid distribution that relies largely on high-density lipoprotein (HDL). These findings provide insights into the genetics and evolution of viral infectivity and cholesterol transport mechanisms in vertebrates.
Collapse
|
34
|
Khwatenge CN, Nahashon SN. Recent Advances in the Application of CRISPR/Cas9 Gene Editing System in Poultry Species. Front Genet 2021; 12:627714. [PMID: 33679892 PMCID: PMC7933658 DOI: 10.3389/fgene.2021.627714] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/19/2021] [Indexed: 12/28/2022] Open
Abstract
CRISPR/Cas9 system genome editing is revolutionizing genetics research in a wide spectrum of animal models in the genetic era. Among these animals, is the poultry species. CRISPR technology is the newest and most advanced gene-editing tool that allows researchers to modify and alter gene functions for transcriptional regulation, gene targeting, epigenetic modification, gene therapy, and drug delivery in the animal genome. The applicability of the CRISPR/Cas9 system in gene editing and modification of genomes in the avian species is still emerging. Up to date, substantial progress in using CRISPR/Cas9 technology has been made in only two poultry species (chicken and quail), with chicken taking the lead. There have been major recent advances in the modification of the avian genome through their germ cell lineages. In the poultry industry, breeders and producers can utilize CRISPR-mediated approaches to enhance the many required genetic variations towards the poultry population that are absent in a given poultry flock. Thus, CRISPR allows the benefit of accessing genetic characteristics that cannot otherwise be used for poultry production. Therefore CRISPR/Cas9 becomes a very powerful and robust tool for editing genes that allow for the introduction or regulation of genetic information in poultry genomes. However, the CRISPR/Cas9 technology has several limitations that need to be addressed to enhance its use in the poultry industry. This review evaluates and provides a summary of recent advances in applying CRISPR/Cas9 gene editing technology in poultry research and explores its potential use in advancing poultry breeding and production with a major focus on chicken and quail. This could aid future advancements in the use of CRISPR technology to improve poultry production.
Collapse
Affiliation(s)
- Collins N. Khwatenge
- Department of Biological Sciences, Tennessee State University, Nashville, IN, United States
- Department of Agriculture and Environmental Sciences, Tennessee State University, Nashville, TN, United States
| | - Samuel N. Nahashon
- Department of Agriculture and Environmental Sciences, Tennessee State University, Nashville, TN, United States
| |
Collapse
|
35
|
Wen J, Wu J, Cao T, Zhi S, Chen Y, Aagaard L, Zhen P, Huang Y, Zhong J, Huang J. Methylation silencing and reactivation of exogenous genes in lentivirus-mediated transgenic mice. Transgenic Res 2021; 30:63-76. [PMID: 33394315 DOI: 10.1007/s11248-020-00224-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 11/24/2020] [Indexed: 12/27/2022]
Abstract
Taking advantage of their ability to integrate their genomes into the host genome, lentiviruses have been used to rapidly produce transgenic mice in biomedical research. In most cases, transgenes delivered by lentiviral vectors have resisted silencing mediated by epigenetic modifications in mice. However, some studies revealed that methylation caused decreased transgene expression in mice. Therefore, there is conflicting evidence regarding the methylation-induced silencing of transgenes delivered by lentiviral transduction in mice. In this study, we present evidence that the human TTR transgene was silenced by DNA methylation in the liver of a transgenic mouse model generated by lentiviral transduction. The density of methylation on the transgene was increased during reproduction, and the expression of the transgene was completely silenced in mice of the F2 generation. Interestingly, 5-azacytidine (5-AzaC), a methyltransferase inhibitor, potently reactivated the silenced genes in neonatal mice whose hepatocytes were actively proliferating and led to stable transgene expression during development. However, 5-AzaC did not rescue liver transgene expression when administered to adult mice. Moreover, 5-AzaC at the given dose had low developmental toxicity in the newborn mice. In summary, we demonstrate the methylation-induced silencing of an exogenous gene in the liver of a mouse model generated by lentiviral transduction and show that the silenced transgene can be safely and efficiently reactivated by 5-AzaC treatment, providing an alternative way to obtain progeny with stable transgene expression in the case of the methylation of exogenous genes in transgenic mice generated by lentiviral transduction.
Collapse
Affiliation(s)
- Jinkun Wen
- Department of Neurology, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen, 529030, China.,MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Jinni Wu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Tianqi Cao
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Shengyao Zhi
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Yuxi Chen
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Lars Aagaard
- Department of Biomedicine, Aarhus University, 8000, Aarhus C, Denmark
| | - Peilin Zhen
- Department of Infectious Diseases, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen, 529030, China
| | - Yanming Huang
- Clinical Experimental Center, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen, 529030, China
| | - Jianxin Zhong
- Department of Neurology, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen, 529030, China
| | - Junjiu Huang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China. .,Clinical Experimental Center, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen, 529030, China. .,Key Laboratory of Reproductive Medicine of Guangdong Province, First Affiliated Hospital and School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China.
| |
Collapse
|
36
|
Chojnacka-Puchta L, Sawicka D. CRISPR/Cas9 gene editing in a chicken model: current approaches and applications. J Appl Genet 2020; 61:221-229. [PMID: 31925767 PMCID: PMC7148258 DOI: 10.1007/s13353-020-00537-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Improvements in genome editing technology in birds using primordial germ cells (PGCs) have made the development of innovative era genome-edited avian models possible, including specific chicken bioreactors, production of knock-in/out chickens, low-allergenicity eggs, and disease-resistance models. New strategies, including CRISPR/Cas9, have made gene editing easy and highly efficient in comparison to the well-known process of homologous recombination. The clustered regularly interspaced short palindromic repeats (CRISPR) technique enables us to understand the function of genes and/or to modify the animal phenotype to fit a specific scientific or production target. To facilitate chicken genome engineering applications, we present a concise description of the method and current application of the CRISPR/Cas9 system in chickens. Different strategies for delivering sgRNAs and the Cas9 protein, we also present extensively. Furthermore, we describe a new gesicle technology as a way to deliver Cas9/sgRNA complexes into target cells, and we discuss the advantages and describe basal applications of the CRISPR/Cas9 system in a chicken model.
Collapse
Affiliation(s)
- Luiza Chojnacka-Puchta
- Department of Bioengineering, Lukasiewicz Research Network, Institute of Biotechnology and Antibiotics, Staroscinska 5, 02-516, Warsaw, Poland. .,Department of Chemical, Biological and Aerosol Hazards, Central Institute for Labour Protection-National Research Institute, Czerniakowska 16, 00-701, Warsaw, Poland.
| | - Dorota Sawicka
- Department of Bioengineering, Lukasiewicz Research Network, Institute of Biotechnology and Antibiotics, Staroscinska 5, 02-516, Warsaw, Poland.,Department of Chemical, Biological and Aerosol Hazards, Central Institute for Labour Protection-National Research Institute, Czerniakowska 16, 00-701, Warsaw, Poland
| |
Collapse
|
37
|
Desaulniers AT, Cederberg RA, Carreiro EP, Gurumurthy CB, White BR. A transgenic pig model expressing a CMV-ZsGreen1 reporter across an extensive array of tissues. J Biomed Res 2020; 35:163-173. [PMID: 33797416 PMCID: PMC8038527 DOI: 10.7555/jbr.34.20200111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Since genetic engineering of pigs can benefit both biomedicine and agriculture, selecting a suitable gene promoter is critically important. The cytomegalovirus (CMV) promoter, which can robustly drive ubiquitous transgene expression, is commonly used at present, yet recent reports suggest tissue-specific activity in the pig. The objective of this study was to quantify ZsGreen1 protein (in lieu of CMV promoter activity) in tissues from pigs harboring a CMV-ZsGreen1 transgene with a single integration site. Tissue samples (n=35) were collected from neonatal hemizygous (n=3) and homozygous (n=3) piglets and ZsGreen1 abundance was determined via immunoblotting. ZsGreen1 was detected in all tissues, except hypothalamus, kidney cortex and oviduct. The expression patterns of homozygous and hemizygous piglets were similar (P>0.05). However, quantification revealed that ZsGreen1 protein levels were tissue-specific. Within neural/endocrine tissues, ZsGreen1 abundance was highest in the anterior pituitary gland, intermediate in the cerebellum and lowest in the cerebrum, spinal cord and posterior pituitary (P<0.05). In the digestive system, ZsGreen1 was more abundant in the salivary gland than esophagus, stomach, pancreas, duodenum, jejunum, ileum, spleen, colon, gallbladder and liver (P<0.05). Interestingly, ZsGreen1 amounts also differed within an organ (i.e., the right ventricle had 3-fold higher levels than the other heart chambers; P<0.05). These results provide useful information for the use of the CMV promoter to drive transgene expression in the pig. Moreover, this swine model represents a novel resource of ZsGreen1-labeled organs and a valuable tool to advance genome editing research.
Collapse
Affiliation(s)
- Amy T Desaulniers
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE 68583-0908, USA
| | - Rebecca A Cederberg
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE 68583-0908, USA
| | - Elizabeth P Carreiro
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE 68583-0908, USA
| | - Channabasavaiah B Gurumurthy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930, USA
| | - Brett R White
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE 68583-0908, USA
| |
Collapse
|
38
|
Yang H, Lee BR, Lee HC, Jung SK, Kim JY, No J, Shanmugam S, Jo YJ, Lee H, Hwang S, Byun SJ. Isolation and characterization of cultured chicken oviduct epithelial cells and in vitro validation of constructed ovalbumin promoter in these cells. Anim Biosci 2020; 34:1321-1330. [PMID: 33332940 PMCID: PMC8255889 DOI: 10.5713/ab.20.0627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 12/07/2020] [Indexed: 01/29/2023] Open
Abstract
Objective Transgenic hens hold a great promise to produce various valuable proteins. Through virus transduction into stage X embryo, the transgene expression under the control of constructed chicken ovalbumin promoters has been successfully achieved. However, a validation system that can evaluate differently developed ovalbumin promoters in in vitro, remains to be developed. Methods In the present study, chicken oviduct epithelial cells (cOECs) were isolated from oviduct tissue and shortly cultured with keratinocyte complete medium supplemented with chicken serum. The isolated cells were characterized with immunofluorescence, western blot, and flow cytometry using oviduct-specific marker. Chicken mutated ovalbumin promoter (Mut-4.4-kb-pOV) was validated in these cells using luciferase reporter analysis. Results The isolated cOECs revealed that the oviduct-specific marker, ovalbumin protein, was clearly detected by immunofluorescence, western blot, and flow cytometry analysis revealed that approximately 79.40% of the cells contained this protein. Also, luciferase reporter analysis showed that the constructed Mut-4.4-kb-pOV exhibited 7.1-fold (p<0.001) higher activity in the cOECs. Conclusion Collectively, these results demonstrate the efficient isolation and characterization of cOECs and validate the activity of the constructed ovalbumin promoter in the cultured cOECs. The in vitro validation of the recombinant promoter activity in cOECs can facilitate the production of efficient transgenic chickens for potential use as bioreactors.
Collapse
Affiliation(s)
- Hyeon Yang
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju 55365, Korea
| | - Bo Ram Lee
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju 55365, Korea
| | - Hwi-Cheul Lee
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju 55365, Korea
| | - Sun Keun Jung
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju 55365, Korea
| | - Ji-Youn Kim
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju 55365, Korea
| | - Jingu No
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju 55365, Korea
| | - Sureshkumar Shanmugam
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju 55365, Korea
| | - Yong Jin Jo
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju 55365, Korea
| | - Haesun Lee
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju 55365, Korea
| | - Seongsoo Hwang
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju 55365, Korea
| | - Sung June Byun
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju 55365, Korea
| |
Collapse
|
39
|
Nagy N, Guyer RA, Hotta R, Zhang D, Newgreen DF, Halasy V, Kovacs T, Goldstein AM. RET overactivation leads to concurrent Hirschsprung disease and intestinal ganglioneuromas. Development 2020; 147:dev190900. [PMID: 32994173 PMCID: PMC7657479 DOI: 10.1242/dev.190900] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 09/08/2020] [Indexed: 12/17/2022]
Abstract
Appropriately balanced RET signaling is of crucial importance during embryonic neural crest cell migration, proliferation and differentiation. RET deficiency, for example, leads to intestinal aganglionosis (Hirschsprung disease), whereas overactive RET can lead to multiple endocrine neoplasia (MEN) syndromes. Some RET mutations are associated with both intestinal aganglionosis and MEN-associated tumors. This seemingly paradoxical occurrence has led to speculation of a 'Janus mutation' in RET that causes overactivation or impairment of RET activity depending on the cellular context. Using an intestinal catenary culture system to test the effects of GDNF-mediated RET activation, we demonstrate the concurrent development of distal colonic aganglionosis and intestinal ganglioneuromas. Interestingly, the tumors induced by GDNF stimulation contain enteric neuronal progenitors capable of reconstituting an enteric nervous system when transplanted into a normal developmental environment. These results suggest that a Janus mutation may not be required to explain co-existing Hirschsprung disease and MEN-associated tumors, but rather that RET overstimulation alone is enough to cause both phenotypes. The results also suggest that reprogramming tumor cells toward non-pathological fates may represent a possible therapeutic avenue for MEN-associated neoplasms.
Collapse
Affiliation(s)
- Nandor Nagy
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, 1094, Hungary
| | - Richard A Guyer
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Ryo Hotta
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Dongcheng Zhang
- Neural Crest Group, Murdoch Children's Research Institute, Parkville, VIC 3052, Australia
| | - Donald F Newgreen
- Neural Crest Group, Murdoch Children's Research Institute, Parkville, VIC 3052, Australia
| | - Viktoria Halasy
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, 1094, Hungary
| | - Tamas Kovacs
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, 1094, Hungary
| | - Allan M Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
40
|
Darbey A, Rebourcet D, Curley M, Kilcoyne K, Jeffery N, Reed N, Milne L, Roesl C, Brown P, Smith LB. A comparison of in vivo viral targeting systems identifies adeno-associated virus serotype 9 (AAV9) as an effective vector for genetic manipulation of Leydig cells in adult mice. Andrology 2020; 9:460-473. [PMID: 32996275 DOI: 10.1111/andr.12915] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/01/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Despite the increasing popularity of deliverable transgenics, a robust and fully validated method for targeting Leydig cells, capable of delivering long-term transgene expression, is yet to be defined. OBJECTIVES We compared three viral vector systems in terms of their cell targeting specificity, longevity of gene expression and impact on targeted cell types when delivered to the interstitial compartment of the mouse testis. MATERIALS & METHODS We delivered lentiviral, adenoviral and adeno-associated (AAV) viral particles to the interstitial compartment of adult mouse testis. Immunolocalization and stereology were performed to characterize ability of vectors to target and deliver transgenes to Leydig cells. RESULTS Viral vectors utilized in this study were found to specifically target Leydig cells when delivered interstitially. Transgene expression in lentiviral-targeted Leydig cells was detected for 7 days post-injection before Leydig cells underwent apoptosis. Adenoviral-delivered transgene expression was detected for 10 days post-injection with no evidence of targeted cell apoptosis. We found serotype differences in AAV injected testis with AAV serotype 9 targeting a significant proportion of Leydig cells. Targeting efficiency increased to an average of 59.63% (and a maximum of 80%) of Leydig cells with the addition of neuraminidase during injection. In AAV injected testis sections, transgene expression was detectable for up to 50 days post-injection. DISCUSSION & CONCLUSION Lentivirus, Adenovirus and Adeno-Associated virus delivery to the testis resulted in key variances in targeting efficiency of Leydig cells and in longevity of transgene expression, but identified AAV9 + Neuraminidase as an efficient vector system for transgene delivery and long-term expression. Simple viral delivery procedures and the commercial availability of viral vectors suggests AAV9 + Neuraminidase will be of significant utility to researchers investigating the genetics underpinning Leydig cell function and holds promise to inform the development of novel therapeutics for the treatment of male reproductive disorders.
Collapse
Affiliation(s)
- Annalucia Darbey
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK.,School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, Australia
| | - Diane Rebourcet
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, Australia
| | - Michael Curley
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Karen Kilcoyne
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Nathan Jeffery
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Natalie Reed
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, Australia
| | - Laura Milne
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Cornelia Roesl
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Pamela Brown
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Lee B Smith
- MRC Centre for Reproductive Health, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK.,School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
41
|
Chu D, Nguyen A, Smith SS, Vavrušová Z, Schneider RA. Stable integration of an optimized inducible promoter system enables spatiotemporal control of gene expression throughout avian development. Biol Open 2020; 9:bio055343. [PMID: 32917762 PMCID: PMC7561481 DOI: 10.1242/bio.055343] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 08/27/2020] [Indexed: 01/18/2023] Open
Abstract
Precisely altering gene expression is critical for understanding molecular processes of embryogenesis. Although some tools exist for transgene misexpression in developing chick embryos, we have refined and advanced them by simplifying and optimizing constructs for spatiotemporal control. To maintain expression over the entire course of embryonic development we use an enhanced piggyBac transposon system that efficiently integrates sequences into the host genome. We also incorporate a DNA targeting sequence to direct plasmid translocation into the nucleus and a D4Z4 insulator sequence to prevent epigenetic silencing. We designed these constructs to minimize their size and maximize cellular uptake, and to simplify usage by placing all of the integrating sequences on a single plasmid. Following electroporation of stage HH8.5 embryos, our tetracycline-inducible promoter construct produces robust transgene expression in the presence of doxycycline at any point during embryonic development in ovo or in culture. Moreover, expression levels can be modulated by titrating doxycycline concentrations and spatial control can be achieved using beads or gels. Thus, we have generated a novel, sensitive, tunable, and stable inducible-promoter system for high-resolution gene manipulation in vivo.
Collapse
Affiliation(s)
- Daniel Chu
- Department of Orthopaedic Surgery, University of California at San Francisco, 513 Parnassus Avenue, S-1164, San Francisco, CA 94143-0514, USA
| | - An Nguyen
- Department of Orthopaedic Surgery, University of California at San Francisco, 513 Parnassus Avenue, S-1164, San Francisco, CA 94143-0514, USA
| | - Spenser S Smith
- Department of Orthopaedic Surgery, University of California at San Francisco, 513 Parnassus Avenue, S-1164, San Francisco, CA 94143-0514, USA
| | - Zuzana Vavrušová
- Department of Orthopaedic Surgery, University of California at San Francisco, 513 Parnassus Avenue, S-1164, San Francisco, CA 94143-0514, USA
| | - Richard A Schneider
- Department of Orthopaedic Surgery, University of California at San Francisco, 513 Parnassus Avenue, S-1164, San Francisco, CA 94143-0514, USA
| |
Collapse
|
42
|
Hagihara Y, Okuzaki Y, Matsubayashi K, Kaneoka H, Suzuki T, Iijima S, Nishijima KI. Primordial germ cell-specific expression of eGFP in transgenic chickens. Genesis 2020; 58:e23388. [PMID: 32776392 DOI: 10.1002/dvg.23388] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/03/2020] [Accepted: 07/07/2020] [Indexed: 11/09/2022]
Abstract
PR domain zinc finger protein 14 (PRDM14) plays an essential role in the development of primordial germ cells (PGCs) in mice. However, its functions in avian species remain unclear. In the present study, we used CRISPR/Cas9 to edit the PRDM14 locus in chickens in order to demonstrate its importance in development. The eGFP gene was introduced into the PRDM14 locus of cultured chicken PGCs to knockout PRDM14 and label PGCs. Chimeric chickens were established by a direct injection of eGFP knocked-in (gene-trapped) PGCs into the blood vessels of Hamburger-Hamilton stages (HH-stages) 13-16 chicken embryos. Gene-trapped chickens were established by crossing a chimeric chicken with a wild-type hen with very high efficiency. Heterozygous gene-trapped chickens grew normally and SSEA-1-positive cells expressed eGFP during HH-stages 13-30. These results indicated the specific expression of eGFP within circulating PGCs and gonadal PGCs. At the blastodermal stage, the ratio of homozygous gene-trapped embryos obtained by crossing heterozygous gene-trapped roosters and hens was almost normal; however, all embryos died soon afterward, suggesting the important roles of PRDM14 in chicken early development.
Collapse
Affiliation(s)
- Yota Hagihara
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Japan
| | - Yuya Okuzaki
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Japan.,Avian Bioscience Research Center, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Kazuma Matsubayashi
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Japan
| | - Hidenori Kaneoka
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Japan
| | - Takayuki Suzuki
- Avian Bioscience Research Center, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan.,Laboratory of Avian Bioscience, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Shinji Iijima
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Japan
| | - Ken-Ichi Nishijima
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Nagoya, Japan.,Avian Bioscience Research Center, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan.,Laboratory of Avian Bioscience, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| |
Collapse
|
43
|
Audet JN. Neurobiological and Ecological Correlates of Avian Innovation. Integr Comp Biol 2020; 60:955-966. [PMID: 32681794 DOI: 10.1093/icb/icaa107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In the wild, particularly in rapidly changing conditions, being capable of solving new problems can increase an animal's chances of survival and reproduction. In the current context of widespread habitat destruction and increasing urbanization, innovativeness might be a crucial trait. In the past few decades, birds have proven to be a model taxon for the study of innovation, thanks to the abundant literature on avian innovation reports. Innovation databases in birds have been successfully employed to assess associations between innovativeness and other traits such as invasion success, life history, generalism, and brain encephalization. In order to more directly assess the causes of variation in innovation, a complementary approach consists in measuring innovativeness in wild-caught animals using problem-solving tasks that mimic innovations in the field. This method can allow for finer scale evaluation of ecological and neural correlates of innovation. Here, I review some of the most important findings on the correlates of innovation, with a particular focus on neural ones. I conclude by discussing avenues for future research, which I suggest should focus on neurobiology.
Collapse
|
44
|
Nagy N, Busalt F, Halasy V, Kohn M, Schmieder S, Fejszak N, Kaspers B, Härtle S. In and Out of the Bursa-The Role of CXCR4 in Chicken B Cell Development. Front Immunol 2020; 11:1468. [PMID: 32765509 PMCID: PMC7381227 DOI: 10.3389/fimmu.2020.01468] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 06/05/2020] [Indexed: 11/13/2022] Open
Abstract
In contrast to mammals, early B cell differentiation and diversification of the antibody repertoire in chickens do not take place in the bone marrow but in a specialized gut associated lymphoid tissue (GALT), the bursa of Fabricius. During embryonic development, B cell precursors migrate to the bursa anlage, where they proliferate and diversify their B cell receptor repertoire. Around hatch these diversified B cells start to emigrate from the bursa of Fabricius and populate peripheral lymphoid organs, but very little is known how the migratory processes are regulated. As CXCL12 (syn. SDF-1) and CXCR4 were shown to be essential for the control of B cell migration during the development of lymphoid tissues in mammals, we analyzed expression and function of this chemokine/chemokine-receptor pair in the chicken bursa. We found a strong variation of mRNA abundance of CXCL12 and CXCR4 in different stages of bursa development, with high abundance of CXCL12 mRNA in the bursa anlage at embryonic day 10 (ED10). In situ hybridization demonstrated disseminated CXCL12 expression in the early bursa anlage, which condensed in the developing follicles and was mainly restricted to the follicle cortex post-hatch. Flow cytometric analysis detected CXCR4 protein already on early B cell stages, increasing during bursal development. Post-hatch, a subpopulation with the hallmarks of emigrating B cells became detectable, which had lower CXCR4 expression, suggesting that downregulation of CXCR4 is necessary to leave the CXCL12-high bursal environment. In vivo blockade of CXCR4 using AMD3100 at the time of B cell precursor immigration strongly inhibited follicle development, demonstrating that CXCL12 attracts pre-bursal B cells into the bursal anlage. Altogether, we show that CXCL12 and its receptor CXCR4 are important for both populating the bursa with B cells and emigration of mature B cells into the periphery post hatch, and that CXCR4 function in primary B cell organs is conserved between mammals and birds.
Collapse
Affiliation(s)
- Nandor Nagy
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Florian Busalt
- Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Viktoria Halasy
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Marina Kohn
- Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Stefan Schmieder
- Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Nora Fejszak
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Bernd Kaspers
- Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Sonja Härtle
- Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
45
|
Serralbo O, Salgado D, Véron N, Cooper C, Dejardin MJ, Doran T, Gros J, Marcelle C. Transgenesis and web resources in quail. eLife 2020; 9:56312. [PMID: 32459172 PMCID: PMC7286689 DOI: 10.7554/elife.56312] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/26/2020] [Indexed: 12/26/2022] Open
Abstract
Due to its amenability to manipulations, to live observation and its striking similarities to mammals, the chicken embryo has been one of the major animal models in biomedical research. Although it is technically possible to genome-edit the chicken, its long generation time (6 months to sexual maturity) makes it an impractical lab model and has prevented it widespread use in research. The Japanese quail (Coturnix coturnix japonica) is an attractive alternative, very similar to the chicken, but with the decisive asset of a much shorter generation time (1.5 months). In recent years, transgenic quail lines have been described. Most of them were generated using replication-deficient lentiviruses, a technique that presents diverse limitations. Here, we introduce a novel technology to perform transgenesis in quail, based on the in vivo transfection of plasmids in circulating Primordial Germ Cells (PGCs). This technique is simple, efficient and allows using the infinite variety of genome engineering approaches developed in other models. Furthermore, we present a website centralizing quail genomic and technological information to facilitate the design of genome-editing strategies, showcase the past and future transgenic quail lines and foster collaborative work within the avian community.
Collapse
Affiliation(s)
- Olivier Serralbo
- Australian Regenerative Medicine Institute (ARMI), Monash University, Clayton, Australia
| | - David Salgado
- Marseille Medical Genetics (GMGF), Aix Marseille University, Marseille, France
| | - Nadège Véron
- Australian Regenerative Medicine Institute (ARMI), Monash University, Clayton, Australia
| | - Caitlin Cooper
- CSIRO Health & Biosecurity, Australian Animal Health Laboratory, Geelong, Australia
| | | | - Timothy Doran
- CSIRO Health & Biosecurity, Australian Animal Health Laboratory, Geelong, Australia
| | - Jérome Gros
- Department of Developmental and Stem Cell Biology, Pasteur Institute, Paris, France
| | - Christophe Marcelle
- Australian Regenerative Medicine Institute (ARMI), Monash University, Clayton, Australia.,Institut NeuroMyoGène (INMG), University Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
46
|
Development of a transposon-based technology for transfection of day 0 chicken embryos. Gene 2020; 730:144318. [DOI: 10.1016/j.gene.2019.144318] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/18/2019] [Accepted: 12/20/2019] [Indexed: 12/12/2022]
|
47
|
Abstract
Animal husbandry is believed to predate farming of crops, and remains a core component of most agricultural systems. Historic breeding strategies were based largely on visual observation, crossing animals that were perceived to display enhanced merit. Advances in sequencing capacity coupled with reduced costs have allowed genomic selection tools to deliver significant contribution to breeding regimes. The application of genome editors to make specific changes to livestock genomes has the potential to deliver additional benefits.
Collapse
Affiliation(s)
- Simon Lillico
- The Roslin Institute and R(D)SVS, The University of Edinburgh, Easter Bush Campus, Roslin, Midlothian, EH25 9RG, UK.
| |
Collapse
|
48
|
Moreno-Jiménez I, Lanham SA, Kanczler JM, Hulsart-Billstrom G, Evans ND, Oreffo ROC. Remodelling of human bone on the chorioallantoic membrane of the chicken egg: De novo bone formation and resorption. J Tissue Eng Regen Med 2019; 12:1877-1890. [PMID: 29893478 DOI: 10.1002/term.2711] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 05/08/2018] [Accepted: 06/01/2018] [Indexed: 12/11/2022]
Abstract
Traditionally used as an angiogenic assay, the chorioallantoic membrane (CAM) assay of the chick embryo offers significant potential as an in vivo model for xenograft organ culture. Viable human bone can be cultivated on the CAM and increases in bone volume are evident; however, it remains unclear by what mechanism this change occurs and whether this reflects the physiological process of bone remodelling. In this study we tested the hypothesis that CAM-induced bone remodelling is a consequence of host and graft mediated processes. Bone cylinders harvested from femoral heads post surgery were placed on the CAM of green fluorescent protein (GFP)-chick embryos for 9 days, followed by micro computed tomography (μCT) and histological analysis. Three-dimensional registration of consecutive μCT-scans showed newly mineralised tissue in CAM-implanted bone cylinders, as well as new osteoid deposition histologically. Immunohistochemistry demonstrated the presence of bone resorption and formation markers (Cathepsin K, SOX9 and RUNX2) co-localising with GFP staining, expressed by avian cells only. To investigate the role of the human cells in the process of bone formation, decellularised bone cylinders were implanted on the CAM and comparable increases in bone volume were observed, indicating that avian cells were responsible for the bone mineralisation process. Finally, CAM-implantation of acellular collagen sponges, containing bone morphogenetic protein 2, resulted in the deposition of extracellular matrix and tissue mineralisation. These studies indicate that the CAM can respond to osteogenic stimuli and support formation or resorption of implanted human bone, providing a humanised CAM model for regenerative medicine research and a novel short-term in vivo model for tissue engineering and biomaterial testing.
Collapse
Affiliation(s)
- Inés Moreno-Jiménez
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Institute of Developmental Sciences University of Southampton, Southampton, UK
| | - Stuart A Lanham
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Institute of Developmental Sciences University of Southampton, Southampton, UK
| | - Janos M Kanczler
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Institute of Developmental Sciences University of Southampton, Southampton, UK
| | - Gry Hulsart-Billstrom
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Institute of Developmental Sciences University of Southampton, Southampton, UK
| | - Nicholas D Evans
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Institute of Developmental Sciences University of Southampton, Southampton, UK
| | - Richard O C Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Institute of Developmental Sciences University of Southampton, Southampton, UK
| |
Collapse
|
49
|
In vivo generation of haematopoietic stem/progenitor cells from bone marrow-derived haemogenic endothelium. Nat Cell Biol 2019; 21:1334-1345. [PMID: 31685991 DOI: 10.1038/s41556-019-0410-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 09/23/2019] [Indexed: 01/22/2023]
Abstract
It is well established that haematopoietic stem and progenitor cells (HSPCs) are generated from a transient subset of specialized endothelial cells termed haemogenic, present in the yolk sac, placenta and aorta, through an endothelial-to-haematopoietic transition (EHT). HSPC generation via EHT is thought to be restricted to the early stages of development. By using experimental embryology and genetic approaches in birds and mice, respectively, we document here the discovery of a bone marrow haemogenic endothelium in the late fetus/young adult. These cells are capable of de novo producing a cohort of HSPCs in situ that harbour a very specific molecular signature close to that of aortic endothelial cells undergoing EHT or their immediate progenies, i.e., recently emerged HSPCs. Taken together, our results reveal that HSPCs can be generated de novo past embryonic stages. Understanding the molecular events controlling this production will be critical for devising innovative therapies.
Collapse
|
50
|
Young JJ, Grayson P, Edwards SV, Tabin CJ. Attenuated Fgf Signaling Underlies the Forelimb Heterochrony in the Emu Dromaius novaehollandiae. Curr Biol 2019; 29:3681-3691.e5. [PMID: 31668620 DOI: 10.1016/j.cub.2019.09.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 08/21/2019] [Accepted: 09/06/2019] [Indexed: 11/19/2022]
Abstract
Powered flight was fundamental to the establishment and radiation of birds. However, flight has been lost multiple times throughout avian evolution. Convergent losses of flight within the ratites (flightless paleognaths, including the emu and ostrich) often coincide with reduced wings. Although there is a wealth of anatomical knowledge for several ratites, the genetic mechanisms causing these changes remain debated. Here, we use a multidisciplinary approach employing embryological, genetic, and genomic techniques to interrogate the mechanisms underlying forelimb heterochrony in emu embryos. We show that the initiation of limb formation, an epithelial to mesenchymal transition (EMT) in the lateral plate mesoderm (LPM) and myoblast migration into the LPM, occur at equivalent stages in the emu and chick. However, the emu forelimb fails to subsequently proliferate. The unique emu forelimb expression of Nkx2.5, previously associated with diminished wing development, initiates after this stage (concomitant with myoblast migration into the LPM) and is therefore unlikely to cause this developmental delay. In contrast, RNA sequencing of limb tissue reveals significantly lower Fgf10 expression in the emu forelimb. Artificially increasing Fgf10 expression in the emu LPM induces ectodermal Fgf8 expression and a limb bud. Analyzing open chromatin reveals differentially active regulatory elements near Fgf10 and Sall-1 in the emu wing, and the Sall-1 enhancer activity is dependent on a likely Fgf-mediated Ets transcription factor-binding site. Taken together, our results suggest that regulatory changes result in lower expression of Fgf10 and a concomitant failure to express genes required for limb proliferation in the early emu wing bud.
Collapse
Affiliation(s)
- John J Young
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Phil Grayson
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA
| | - Scott V Edwards
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA
| | - Clifford J Tabin
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|