1
|
Lin Q, Takebayashi K, Torigoe N, Liu B, Namula Z, Hirata M, Tanihara F, Nagahara M, Otoi T. Genome editing of porcine zygotes via lipofection of two guide RNAs using a CRISPR/Cas9 system. J Reprod Dev 2024; 70:356-361. [PMID: 39218670 DOI: 10.1262/jrd.2024-054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
CRISPR/Cas9-based multiplex genome editing via electroporation is relatively efficient; however, lipofection is versatile because of its ease of use and low cost. Here, we aimed to determine the efficiency of lipofection in CRISPR/Cas9-based multiplex genome editing using growth hormone receptor (GHR) and glycoprotein alpha-galactosyltransferase 1 (GGTA1)-targeting guide RNAs (gRNAs) in pig zygotes. Zona pellucida-free zygotes were collected 10 h after in vitro fertilization and incubated with Cas9, gRNAs, and Lipofectamine 2000 (LP2000) for 5 h. In Experiment 1, we evaluated the mutation efficiency of gRNAs targeting either GHR or GGTA1 in zygotes transfected using LP2000 and cultured in 4-well plates. In Experiment 2, we examined the effects of the culture method on the development, mutation rate, and mutation efficiency of zygotes with simultaneously double-edited GHR and GGTA1, cultured using 4-well (group culture) and 25-well plates (individual culture). In Experiment 3, we assessed the effect of additional GHR-targeted lipofection before and after simultaneous double gRNA-targeted lipofection on the mutation efficiency of edited embryos cultured in 25-well plates. No significant differences in mutation rates were observed between the zygotes edited with either gRNA. Moreover, the formation rate of blastocysts derived from GHR and GGTA1 double-edited zygotes was significantly increased in the 25-well plate culture compared to that in the 4-well plate culture. However, mutations were only observed in GGTA1 when zygotes were transfected with both gRNAs, irrespective of the culture method used. GHR mutations were detected only in blastocysts derived from zygotes subjected to GHR-targeted lipofection before simultaneous double gRNA-targeted lipofection. Overall, our results suggest that additional lipofection before simultaneous double gRNA-targeted lipofection induces additional mutations in the zygotes.
Collapse
Affiliation(s)
- Qingyi Lin
- Bio-Innovation Research Center, Tokushima University, Tokushima 779-3233, Japan
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima 779-3233, Japan
| | - Koki Takebayashi
- Bio-Innovation Research Center, Tokushima University, Tokushima 779-3233, Japan
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima 779-3233, Japan
| | - Nanaka Torigoe
- Bio-Innovation Research Center, Tokushima University, Tokushima 779-3233, Japan
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima 779-3233, Japan
| | - Bin Liu
- Bio-Innovation Research Center, Tokushima University, Tokushima 779-3233, Japan
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima 779-3233, Japan
| | - Zhao Namula
- Bio-Innovation Research Center, Tokushima University, Tokushima 779-3233, Japan
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524091, China
| | - Maki Hirata
- Bio-Innovation Research Center, Tokushima University, Tokushima 779-3233, Japan
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima 779-3233, Japan
| | - Fuminori Tanihara
- Center for Development of Advanced Medical Technology, Jichi Medical University, Tochigi 329-0498, Japan
| | - Megumi Nagahara
- Bio-Innovation Research Center, Tokushima University, Tokushima 779-3233, Japan
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima 779-3233, Japan
| | - Takeshige Otoi
- Bio-Innovation Research Center, Tokushima University, Tokushima 779-3233, Japan
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima 779-3233, Japan
| |
Collapse
|
2
|
Garcia-Canovas M, Parrilla I, Cuello C, Gil MA, Martinez EA. Swine in vitro embryo production: Potential, challenges, and advances. Anim Reprod Sci 2024; 270:107600. [PMID: 39270509 DOI: 10.1016/j.anireprosci.2024.107600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/15/2024]
Abstract
Pig production, a vital sector of the meat industry, faces demands for improved quality, efficiency, and sustainability. Advancements in breeding, disease control, and artificial insemination have enhanced production, while biotechnologies such as in vitro embryo production (IVP) and genetic engineering offer further progress. In vitro embryo production could facilitate the global exchange of valuable genetic material, accelerate breeding programs, and improve productivity, and it is essential for generating genetically modified (GM) pigs. These GM pigs have two main applications: first, they allow for targeted modifications aimed at improving production traits relevant to pig production in agriculture, such as meat quality and disease resistance. Second, they serve as valuable biomedical models for human disease research, regenerative medicine, and organ transplantation. Yet, despite notable advancements in recent decades, the efficiency of the current IVP systems for porcine embryos remains a challenge. Compared to the in vivo environment, suboptimal culture conditions lead to issues such as elevated polyspermy, poor embryo development, and the production of low-quality blastocysts. This review provides an overview of the key steps and recent advancements in porcine IVP technology. We will emphasize the promising utilization of oocytes from live females of high genetic value through ovum pick-up and the incorporation of extracellular vesicles and cytokines into IVP media. These innovative strategies hold immense potential to significantly enhance embryo development and overall success rates in porcine IVP, and could open the door for significant progress in both agriculture and biomedicine applications.
Collapse
Affiliation(s)
- Manuela Garcia-Canovas
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", Institute for Biomedical Research of Murcia (IMIB-Arrixaca), University of Murcia, Murcia 30100, Spain
| | - Inmaculada Parrilla
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", Institute for Biomedical Research of Murcia (IMIB-Arrixaca), University of Murcia, Murcia 30100, Spain
| | - Cristina Cuello
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", Institute for Biomedical Research of Murcia (IMIB-Arrixaca), University of Murcia, Murcia 30100, Spain
| | - Maria A Gil
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", Institute for Biomedical Research of Murcia (IMIB-Arrixaca), University of Murcia, Murcia 30100, Spain.
| | - Emilio A Martinez
- Department of Medicine and Animal Surgery, Faculty of Veterinary Medicine, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum", Institute for Biomedical Research of Murcia (IMIB-Arrixaca), University of Murcia, Murcia 30100, Spain
| |
Collapse
|
3
|
Lin Q, Takebayashi K, Torigoe N, Liu B, Namula Z, Hirata M, Nagahara M, Tanihara F, Otoi T. Evaluation of culture methods and chemical reagent combinations on CRISPR/Cas9 gene editing systems by lipofection in pig zygotes. In Vitro Cell Dev Biol Anim 2024; 60:725-731. [PMID: 38664280 DOI: 10.1007/s11626-024-00908-0] [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: 02/20/2024] [Accepted: 04/04/2024] [Indexed: 08/03/2024]
Abstract
The delivery of CRISPR/Cas ribonucleoprotein (RNP) complexes is gaining attention owing to its high cleavage efficiency and reduced off-target effects. Although RNPs can be delivered into porcine zygotes via electroporation with relatively high efficiency, lipofection-mediated transfection appears to be versatile because of its ease of use, low cost, and adaptation to high-throughput systems. However, this system requires improvements in terms of embryo development and mutation rates. Therefore, this study elucidated the effects of culture methods and reagent combinations on the CRISPR/Cas9 gene editing systems by using three lipofection reagents: Lipofectamine™ CRISPRMAX™ Cas9 Transfection Reagent (CM), Lipofectamine™ 2000 Transfection Reagent (LP), and jetCRISPR™ RNP Transfection Reagent (Jet). Porcine zona pellucida-free zygotes were incubated for 5 h with Cas9, a guide RNA targeting CD163, and the above lipofection reagents. When examining the effect of culture methods using 4-well (multiple embryo culture) and 25-well plates (single embryo culture) on the efficiency of CM-mediated zygote transfection, the culture of embryos in 25-well plates significantly increased the blastocyst formation rate; however, there was no difference in mutation rates between the 4-well and 25-well plates. When assessing the effects of individual or combined reagents on the efficiency of zygote transfection, the mutation rate was significantly lower for individual LP compared to individual CM- and Jet-mediated transfections. Moreover, combinations of lipofection transfection reagents did not significantly increase the mutation rate or mutation efficiency.
Collapse
Affiliation(s)
- Qingyi Lin
- Laboratory of Animal Reproduction, Bio-Innovation Research Center, Tokushima University, 2272-1 Ishii, Myozai-Gun, Tokushima, 779-3233, Japan
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, 779-3233, Japan
| | - Koki Takebayashi
- Laboratory of Animal Reproduction, Bio-Innovation Research Center, Tokushima University, 2272-1 Ishii, Myozai-Gun, Tokushima, 779-3233, Japan
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, 779-3233, Japan
| | - Nanaka Torigoe
- Laboratory of Animal Reproduction, Bio-Innovation Research Center, Tokushima University, 2272-1 Ishii, Myozai-Gun, Tokushima, 779-3233, Japan
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, 779-3233, Japan
| | - Bin Liu
- Laboratory of Animal Reproduction, Bio-Innovation Research Center, Tokushima University, 2272-1 Ishii, Myozai-Gun, Tokushima, 779-3233, Japan
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, 779-3233, Japan
| | - Zhao Namula
- Laboratory of Animal Reproduction, Bio-Innovation Research Center, Tokushima University, 2272-1 Ishii, Myozai-Gun, Tokushima, 779-3233, Japan
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, China
| | - Maki Hirata
- Laboratory of Animal Reproduction, Bio-Innovation Research Center, Tokushima University, 2272-1 Ishii, Myozai-Gun, Tokushima, 779-3233, Japan
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, 779-3233, Japan
| | - Megumi Nagahara
- Laboratory of Animal Reproduction, Bio-Innovation Research Center, Tokushima University, 2272-1 Ishii, Myozai-Gun, Tokushima, 779-3233, Japan
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, 779-3233, Japan
| | - Fuminori Tanihara
- Laboratory of Animal Reproduction, Bio-Innovation Research Center, Tokushima University, 2272-1 Ishii, Myozai-Gun, Tokushima, 779-3233, Japan
| | - Takeshige Otoi
- Laboratory of Animal Reproduction, Bio-Innovation Research Center, Tokushima University, 2272-1 Ishii, Myozai-Gun, Tokushima, 779-3233, Japan.
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, 779-3233, Japan.
| |
Collapse
|
4
|
He Y, Gao M, Zhu X, Peng W, Zhou Y, Cheng J, Bai L, Bao J. Large-Scale Formation and Long-Term Culture of Hepatocyte Organoids From Streamlined In Vivo Genome-Edited GGTA1 -/- Pigs for Bioartificial Liver Applications. Xenotransplantation 2024; 31:e12878. [PMID: 39166823 DOI: 10.1111/xen.12878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/13/2024] [Accepted: 07/30/2024] [Indexed: 08/23/2024]
Abstract
Hepatocyte transplantation and bioartificial liver (BAL) systems hold significant promise as less invasive alternatives to traditional transplantation, providing crucial temporary support for patients with acute and chronic liver failure. Although human hepatocytes are ideal, their use is limited by ethical concerns and donor availability, leading to the use of porcine hepatocytes in BAL systems due to their functional similarities. Recent advancements in gene-editing technology have improved porcine organ xenotransplantation clinical trials by addressing immune rejection issues. Gene-edited pigs, such as alpha-1,3-galactosyltransferase (GGTA1) knockout pigs, offer a secure source of primary cells for BAL systems. Our research focuses on optimizing the safety and functionality of porcine primary hepatocytes during large-scale cultivation. We achieved this by creating GGTA1 knockout pigs through one-step delivery of CRISPR/Cas9 to pig zygotes via oviduct injection of rAAV, and enhancing hepatocyte viability and function by co-culturing hepatocytes with Roof plate-specific spondin 1 overexpressing HUVECs (R-HUVECs). Using a Rocker culture system, approximately 1010 primary porcine hepatocytes and R-HUVECs rapidly formed organoids with a diameter of 92.1 ± 28.1 µm within 24 h. These organoids not only maintained excellent functionality but also supported partial hepatocyte self-renewal during long-term culture over 28 days. Gene-edited primary porcine hepatocyte organoids will significantly advance the applications of hepatocyte transplantation and BAL systems.
Collapse
Affiliation(s)
- Yuting He
- Department of Pathology, Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Mengyu Gao
- Department of Pathology, Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xinglong Zhu
- Department of Pathology, Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Wanliu Peng
- Department of Pathology, Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yanyan Zhou
- Department of Pathology, Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jingqiu Cheng
- Key Laboratory of Transplant Engineering and Immunology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Lang Bai
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ji Bao
- Department of Pathology, Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
5
|
Navarro-Serna S, Piñeiro-Silva C, Fernández-Martín I, Dehesa-Etxebeste M, López de Munain A, Gadea J. Oocyte electroporation prior to in vitro fertilization is an efficient method to generate single, double, and multiple knockout porcine embryos of interest in biomedicine and animal production. Theriogenology 2024; 218:111-118. [PMID: 38320372 DOI: 10.1016/j.theriogenology.2024.01.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/31/2024] [Accepted: 01/31/2024] [Indexed: 02/08/2024]
Abstract
Genetically modified pigs play a critical role in mimicking human diseases, xenotransplantation, and the development of pigs resistant to viral diseases. The use of programmable endonucleases, including the CRISPR/Cas9 system, has revolutionized the generation of genetically modified pigs. This study evaluates the efficiency of electroporation of oocytes prior to fertilization in generating edited gene embryos for different models. For single gene editing, phospholipase C zeta (PLC ζ) and fused in sarcoma (FUS) genes were used, and the concentration of sgRNA and Cas9 complexes was optimized. The results showed that increasing the concentration resulted in higher mutation rates without affecting the blastocyst rate. Electroporation produced double knockouts for the TPC1/TPC2 genes with high efficiency (79 %). In addition, resistance to viral diseases such as PRRS and swine influenza was achieved by electroporation, allowing the generation of double knockout embryo pigs (63 %). The study also demonstrated the potential for multiple gene editing in a single step using electroporation, which is relevant for xenotransplantation. The technique resulted in the simultaneous mutation of 5 genes (GGTA1, B4GALNT2, pseudo B4GALNT2, CMAH and GHR). Overall, electroporation proved to be an efficient and versatile method to generate genetically modified embryonic pigs, offering significant advances in biomedical and agricultural research, xenotransplantation, and disease resistance. Electroporation led to the processing of numerous oocytes in a single session using less expensive equipment. We confirmed the generation of gene-edited porcine embryos for single, double, or quintuple genes simultaneously without altering embryo development to the blastocyst stage. The results provide valuable insights into the optimization of gene editing protocols for different models, opening new avenues for research and applications in this field.
Collapse
Affiliation(s)
- Sergio Navarro-Serna
- Department Physiology, University of Murcia, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum" and Institute for Biomedical Research of Murcia (IMIB-Arrixaca), 30100, Murcia, Spain
| | - Celia Piñeiro-Silva
- Department Physiology, University of Murcia, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum" and Institute for Biomedical Research of Murcia (IMIB-Arrixaca), 30100, Murcia, Spain
| | - Irene Fernández-Martín
- Department Physiology, University of Murcia, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum" and Institute for Biomedical Research of Murcia (IMIB-Arrixaca), 30100, Murcia, Spain
| | | | - Adolfo López de Munain
- IIS Biodonostia, Neuroscience, San Sebastián, Spain; Department of Neurology. Hospital Universitario Donostia-OSAKIDETZA, San Sebastián, Spain; Department of Neurosciences. University of the Basque Country (UPV-EHU), San Sebastián, Spain; CIBERNED (CIBER), Institute Carlos III, Madrid, Spain
| | - Joaquín Gadea
- Department Physiology, University of Murcia, International Excellence Campus for Higher Education and Research "Campus Mare Nostrum" and Institute for Biomedical Research of Murcia (IMIB-Arrixaca), 30100, Murcia, Spain.
| |
Collapse
|
6
|
Rosenbaum Bartkova A, Nemcova L, Strejcek F, Gad A, Kinterova V, Morovic M, Benc M, Prochazka R, Laurincik J. Impact of media supplements FGF2, LIF and IGF1 on the genome activity of porcine embryos produced in vitro. Sci Rep 2024; 14:7081. [PMID: 38528099 PMCID: PMC10963758 DOI: 10.1038/s41598-024-57865-7] [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: 09/14/2023] [Accepted: 03/22/2024] [Indexed: 03/27/2024] Open
Abstract
In this article, we focused on the impact of precisely chemically modified FLI maturation medium enriched with fibroblast growth factor 2 (FGF2), leukemia inhibitory factor (LIF), insulin-like growth factor 1 (IGF1), and polyvinyl alcohol (PVA) and its potential to improve the efficiency of in vitro production of porcine embryos. We hypothesized that enhancing the composition of the maturation medium could result in an elevated production of embryos in vitro and can affect EGA. FLI medium resulted in a significantly higher rate of oocyte blastocyst maturation and formation compared to the control DMEM medium. In addition, immunocytochemical labelling confirmed the detection of UBF in 4-cell FLI parthenogenic embryos, suggesting similarities with natural embryo development. Through RNAseq analysis, upregulated genes present in 4-cell FLI embryos were found to play key roles in important biological processes such as cell proliferation, cell differentiation, and transcriptional regulation. Based on our findings, we demonstrated the positive influence of FLI medium in the evaluation of in vitro embryo production, EGA detection, transcriptomic and proteomic profile, which was confirmed by the positive activation of the embryonal genome in the 4-cell stage of parthenogenetically activated embryos.
Collapse
Affiliation(s)
- Alexandra Rosenbaum Bartkova
- Constantine the Philosopher University in Nitra, Nitra, Slovakia
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Liběchov, Czech Republic
| | - Lucie Nemcova
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Liběchov, Czech Republic
| | | | - Ahmed Gad
- Animal Reproduction and Biotechnology Laboratory (ARBL), Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
- Department of Animal Production, Faculty of Agriculture, Cairo University, Giza, Egypt
| | - Veronika Kinterova
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Liběchov, Czech Republic
| | - Martin Morovic
- Constantine the Philosopher University in Nitra, Nitra, Slovakia
| | - Michal Benc
- Constantine the Philosopher University in Nitra, Nitra, Slovakia
| | - Radek Prochazka
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Liběchov, Czech Republic
| | - Jozef Laurincik
- Constantine the Philosopher University in Nitra, Nitra, Slovakia
| |
Collapse
|
7
|
Herdiana Y, Sofian FF, Shamsuddin S, Rusdiana T. Towards halal pharmaceutical: Exploring alternatives to animal-based ingredients. Heliyon 2024; 10:e23624. [PMID: 38187251 PMCID: PMC10770512 DOI: 10.1016/j.heliyon.2023.e23624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 01/09/2024] Open
Abstract
Halal is a crucial concept for Muslim consumers regarding consumed products, including pharmaceutical ingredients, which are essential in modern medicine. To address the issue of using porcine-sourced ingredients in pharmaceuticals, it is essential to search for halal alternatives derived from poultry, animal by-products from meat processing, marine sources, and plants. However, the complexity of this problem is further compounded by the rapid advances in innovation and technology, which can lead to adulteration of ingredients derived from pigs. Other challenges include the sustainability of alternative materials, management of waste or by-products practice, halal awareness, certification, government policies, religious adherence of consumers, food suppliers, marketers, and purchasing of products. The importance of halal and non-halal problems, specifically in the context of pharmaceutical materials, is still rarely discussed, including alternatives derived from poultry, animal by-products, marine sources, and plants. Due to the increasing global population, there is a growing need to increase awareness and concern among Muslim consumers for halal products, including pharmaceuticals. Therefore, this research aimed to investigate the importance of halal and non-halal issues in pharmaceutical ingredients, the potential impact on the Muslim community, as well as opportunities and challenges in the search for alternative ingredients.
Collapse
Affiliation(s)
- Yedi Herdiana
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45363, Indonesia
- Halal Food Pharmaceutical and Healthcare Society, Faculty of Pharmacy, Padjadjaran University, Sumedang, 45363, Indonesia
| | - Ferry Ferdiansyah Sofian
- Department of Pharmaceutical Biology, Faculty of Pharmacy, Padjadjaran University, Sumedang, 45363, Indonesia
- Halal Food Pharmaceutical and Healthcare Society, Faculty of Pharmacy, Padjadjaran University, Sumedang, 45363, Indonesia
| | - Shaharum Shamsuddin
- School of Health Sciences, Universiti Sains Malaysia, 16150, Kubang Kerian, Kelantan, Malaysia
- Nanobiotech Research Initiative, Institute for Research in Molecular Medicine (INFORMM), USM, 11800, Penang, Malaysia
- USM-RIKEN Interdisciplinary Collaboration on Advanced Sciences (URICAS), 11800, USM, Penang, Malaysia
| | - Taofik Rusdiana
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45363, Indonesia
| |
Collapse
|
8
|
Somfai T. Vitrification of immature oocytes in pigs. Anim Sci J 2024; 95:e13943. [PMID: 38578008 DOI: 10.1111/asj.13943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/27/2024] [Accepted: 03/04/2024] [Indexed: 04/06/2024]
Abstract
Cryopreservation of oocytes is an important technology for the in vitro gene banking of female germplasm. Although slow freezing is not feasible, porcine oocytes survive vitrification at high rates. Cryopreservation at the germinal vesicle stage appears to be more advantageous than that at the metaphase-II stage. Several factors are considered to affect the success of vitrification and subsequent utilization of immature porcine oocytes such as the device, the protocols for cryoprotectant application, warming, and the post-warming culture. Although live piglets could be obtained from vitrified immature oocytes, their competence to develop to the blastocyst stage is still reduced compared to their non-vitrified counterparts, indicating that there is room for further improvement. Vitrified oocytes suffer various types of damage and alteration which may reduce their developmental ability. Some of these can recover to some extent during subsequent culture, such as the damage of the cytoskeleton and mitochondria. Others such as premature nuclear progression, DNA damage and epigenetic alterations will require further research to be clarified and addressed. To date, the practical application of oocyte vitrification in pigs has been confined to the gene banking of a few native breeds.
Collapse
Affiliation(s)
- Tamás Somfai
- Animal Model Development Group, Division of Biomaterial Sciences, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Japan
| |
Collapse
|
9
|
Okamoto K, Matsunari H, Nakano K, Umeyama K, Hasegawa K, Uchikura A, Takayanagi S, Watanabe M, Ohgane J, Stirm M, Kurome M, Klymiuk N, Nagaya M, Wolf E, Nagashima H. Phenotypic features of genetically modified DMD-X KOX WT pigs. Regen Ther 2023; 24:451-458. [PMID: 37772130 PMCID: PMC10523442 DOI: 10.1016/j.reth.2023.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/22/2023] [Accepted: 09/11/2023] [Indexed: 09/30/2023] Open
Abstract
Introduction Duchenne muscular dystrophy (DMD) is a hereditary neuromuscular disorder caused by mutation in the dystrophin gene (DMD) on the X chromosome. Female DMD carriers occasionally exhibit symptoms such as muscle weakness and heart failure. Here, we investigated the characteristics and representativeness of female DMD carrier (DMD-XKOXWT) pigs as a suitable disease model. Methods In vitro fertilization using sperm from a DMD-XKOY↔XWTXWT chimeric boar yielded DMD-XKOXWT females, which were used to generate F2 and F3 progeny, including DMD-XKOXWT females. F1-F3 piglets were genotyped and subjected to biochemical analysis for blood creatine kinase (CK), aspartate aminotransferase, and lactate dehydrogenase. Skeletal muscle and myocardial tissue were analyzed for the expression of dystrophin and utrophin, as well as for lymphocyte and macrophage infiltration. Results DMD-XKOXWT pigs exhibited various characteristics common to human DMD carrier patients, namely, asymptomatic hyperCKemia, dystrophin expression patterns in the skeletal and cardiac muscles, histopathological features of skeletal muscle degeneration, myocardial lesions in adulthood, and sporadic death. Pathological abnormalities observed in the skeletal muscles in DMD-XKOXWT pigs point to a frequent incidence of pathological abnormalities in the musculoskeletal tissues of latent DMD carriers. Our findings suggest a higher risk of myocardial abnormalities in DMD carrier women than previously believed. Conclusions We demonstrated that DMD-XKOXWT pigs could serve as a suitable large animal model for understanding the pathogenic mechanism in DMD carriers and developing therapies for female DMD carriers.
Collapse
Affiliation(s)
- Kazutoshi Okamoto
- Laboratory of Medical Bioengineering, Department of Life Sciences, School of Agriculture, Meiji University, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
| | - Hitomi Matsunari
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
| | - Kazuaki Nakano
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
| | - Kazuhiro Umeyama
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
| | - Koki Hasegawa
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
| | - Ayuko Uchikura
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
| | - Shuko Takayanagi
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
| | - Masahito Watanabe
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
| | - Jun Ohgane
- Laboratory of Genomic Function Engineering, Department of Life Sciences, School of Agriculture, Meiji University, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
| | - Michael Stirm
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, 81377 Munich, Germany
- Center for Innovative Medical Models (CiMM), LMU Munich, 85764 Oberschleissheim, Germany
| | - Mayuko Kurome
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, 81377 Munich, Germany
- Center for Innovative Medical Models (CiMM), LMU Munich, 85764 Oberschleissheim, Germany
| | - Nikolai Klymiuk
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, 81377 Munich, Germany
- Center for Innovative Medical Models (CiMM), LMU Munich, 85764 Oberschleissheim, Germany
| | - Masaki Nagaya
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
| | - Eckhard Wolf
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, 81377 Munich, Germany
- Center for Innovative Medical Models (CiMM), LMU Munich, 85764 Oberschleissheim, Germany
| | - Hiroshi Nagashima
- Laboratory of Medical Bioengineering, Department of Life Sciences, School of Agriculture, Meiji University, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa 214-8571, Japan
| |
Collapse
|
10
|
Park MR, Ahn JS, Lee MG, Lee BR, Ock SA, Byun SJ, Hwang IS. Characterization of Enlarged Tongues in Cloned Piglets. Curr Issues Mol Biol 2023; 45:9103-9116. [PMID: 37998748 PMCID: PMC10670481 DOI: 10.3390/cimb45110571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/10/2023] [Accepted: 11/13/2023] [Indexed: 11/25/2023] Open
Abstract
Although the efficiency of cloning remains very low, this technique has become the most reliable way to produce transgenic pigs. However, the high rate of abnormal offspring such as an enlarged tongue lowers the cloning efficiency by reducing the early survivability of piglets. Thus, the present study was conducted to identify the characteristics of the enlarged tongue from cloned piglets by histologic and transcriptomic analysis. As a result, it was observed that the tissues from enlarged tongues (n = 3) showed isolated and broken muscle bundles with wide spaces while the tissues from normal tongues (n = 3) showed the tight connection of muscle bundles without space by histological analysis. Additionally, transmission electron microscopy results also showed the formation of isolated and broken muscle bundles in enlarged tongues. The transcriptome analysis showed a total of 197 upregulated and 139 downregulated genes with more than 2-fold changes in enlarged tongues. Moreover, there was clear evidence for the difference between groups in the muscle system process with high relation in the biological process by gene ontology analysis. The analysis of the Kyoto Encyclopedia of Gene and Genomes pathway of differentially expressed genes indicated that the pentose phosphate pathway, glycolysis/gluconeogenesis, and glucagon signaling pathway were also involved. Conclusively, our results could suggest that the abnormal glycolytic regulation may result in the formation of an enlarged tongue. These findings might have the potential to understand the underlying mechanisms, abnormal development, and disease diagnosis in cloned pigs.
Collapse
Affiliation(s)
- Mi-Ryung Park
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju 55365, Republic of Korea; (M.-R.P.)
| | - Jin Seop Ahn
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - Min Gook Lee
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju 55365, Republic of Korea; (M.-R.P.)
| | - Bo Ram Lee
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju 55365, Republic of Korea; (M.-R.P.)
| | - Sun A Ock
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju 55365, Republic of Korea; (M.-R.P.)
| | - Sung June Byun
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju 55365, Republic of Korea; (M.-R.P.)
| | - In-Sul Hwang
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju 55365, Republic of Korea; (M.-R.P.)
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| |
Collapse
|
11
|
Zheng H, Choi H, Oh D, Kim M, Cai L, Jawad A, Kim S, Lee J, Hyun SH. Supplementation with fibroblast growth factor 7 during in vitro maturation of porcine cumulus-oocyte complexes improves oocyte maturation and early embryonic development. Front Vet Sci 2023; 10:1250551. [PMID: 38026656 PMCID: PMC10662523 DOI: 10.3389/fvets.2023.1250551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 10/13/2023] [Indexed: 12/01/2023] Open
Abstract
In vitro generation of porcine embryos is an indispensable method in the realms of both agriculture and biomedicine. Nonetheless, the extant procedures encounter substantial obstacles pertaining to both the caliber and efficacy of the produced embryos, necessitating extensive research to in vitro maturation (IVM), the seminal commencement phase. One potentially fruitful approach may lie in refining the media and supplements composition utilized for oocyte maturation. Fibroblast growth factor-7 (FGF7), alternatively termed keratinocyte growth factor, is a theca-derived cytokine integral to folliculogenesis. This study aimed to examine the ramifications of supplementing FGF7 during the IVM phase. To determine the FGF7 location and its receptor in porcine ovaries, immunohistochemistry was executed based on follicle size categories (1-2, 3-6, and 7-9 mm). Regardless of follicle size, it was determined that FGF7 was expressed in theca and granulosa cells (GCs), whereas the FGF7 receptor was only expressed in the GCs of the larger follicles. During the IVM process, the maturation medium was supplied with various concentrations of FGF7, aiming to mature porcine cumulus-oocyte complexes (COCs). The data indicated a significant augmentation in the nuclear maturation rate only within the group treated with 10 ng/mL of FGF7 (p < 0.05). Post-IVM, the oocytes diameter exhibited a significant expansion in all groups that received FGF7 supplementation (p < 0.05). Additionally, all FGF7-supplemented groups exhibited a substantial elevation in intracellular glutathione levels, coupled with a noticeable reduction in reactive oxygen species levels (p < 0.05). With respect to gene expressions related to apoptosis, FGF7 treatment elicited a downregulation of pro-apoptotic genes and an upregulation of anti-apoptotic genes. The expression of genes associated with antioxidants underwent a significant enhancement (p < 0.05). In terms of the FGF7 signaling pathway-associated genes, there was a significant elevation in the mRNA expression of ERK1, ERK2, c-kit, and KITLG (p < 0.05). Remarkably, the group of 10 ng/mL of FGF7 demonstrated an appreciable uptick in the blastocyst formation rate during embryonic development post-parthenogenetic activation (p < 0.05). In conclusion, the FGF7 supplementation during IVM substantially augments the quality of matured oocytes and facilitates the subsequent development of parthenogenetically activated embryos. These results offer fresh perspectives on improved maturation and following in vitro evolution of porcine oocytes.
Collapse
Affiliation(s)
- Haomiao Zheng
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Research Institute for Natural Science, Chungbuk National University, Cheongju, Republic of Korea
| | - Hyerin Choi
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Research Institute for Natural Science, Chungbuk National University, Cheongju, Republic of Korea
| | - Dongjin Oh
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Research Institute for Natural Science, Chungbuk National University, Cheongju, Republic of Korea
| | - Mirae Kim
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Research Institute for Natural Science, Chungbuk National University, Cheongju, Republic of Korea
| | - Lian Cai
- Department of Biological Sciences, College of Arts and Sciences, University at Buffalo, The State University of New York (SUNY), Buffalo, NY, United States
| | - Ali Jawad
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Research Institute for Natural Science, Chungbuk National University, Cheongju, Republic of Korea
| | - Sohee Kim
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Research Institute for Natural Science, Chungbuk National University, Cheongju, Republic of Korea
| | - Joohyeong Lee
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Research Institute for Natural Science, Chungbuk National University, Cheongju, Republic of Korea
| | - Sang-Hwan Hyun
- Laboratory of Veterinary Embryology and Biotechnology (VETEMBIO), Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
- Institute of Stem Cell and Regenerative Medicine (ISCRM), Research Institute for Natural Science, Chungbuk National University, Cheongju, Republic of Korea
- Graduate School of Veterinary Biosecurity and Protection, Chungbuk National University, Cheongju, Republic of Korea
| |
Collapse
|
12
|
Kim SE, Sun WS, Oh M, Lee S, No JG, Lee H, Lee P, Oh KB. Identification of the Porcine Vascular Endothelial Cell-Specific Promoter ESAM1.0 Using Transcriptome Analysis. Genes (Basel) 2023; 14:1928. [PMID: 37895277 PMCID: PMC10606829 DOI: 10.3390/genes14101928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/06/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
The vascular endothelium of xenografted pig organs represents the initial site of rejection after exposure to recipient immune cells. In this study, we aimed to develop a promoter specific to porcine vascular endothelial cells as a step toward overcoming xenograft rejection. Transcriptome analysis was performed on porcine aortic endothelial cells (PAECs), ear skin fibroblasts isolated from GGTA knockout (GTKO) pigs, and the porcine renal epithelial cell line pk-15. RNA sequencing confirmed 243 differentially expressed genes with expression changes of more than 10-fold among the three cell types. Employing the Human Protein Atlas database as a reference, we identified 34 genes exclusive to GTKO PAECs. The endothelial cell-specific adhesion molecule (ESAM) was selected via qPCR validation and showed high endothelial cell specificity and stable expression across tissues. We selected 1.0 kb upstream sequences of the translation start site of the gene as the promoter ESAM1.0. A luciferase assay revealed that ESAM1.0 promoter transcriptional activity was significant in PAECs, leading to a 2.8-fold higher level of expression than that of the porcine intercellular adhesion molecule 2 (ICAM2) promoter, which is frequently used to target endothelial cells in transgenic pigs. Consequently, ESAM1.0 will enable the generation of genetically modified pigs with endothelium-specific target genes to reduce xenograft rejection.
Collapse
Affiliation(s)
- Sang Eun Kim
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Jeonju-si 55365, Jeollabuk-do, Republic of Korea; (S.E.K.); (W.-S.S.); (M.O.); (S.L.); (J.-G.N.); (H.L.); (P.L.)
| | - Wu-Sheng Sun
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Jeonju-si 55365, Jeollabuk-do, Republic of Korea; (S.E.K.); (W.-S.S.); (M.O.); (S.L.); (J.-G.N.); (H.L.); (P.L.)
- College of Veterinary Medicine, Jilin Agricultural University, Changchun 130118, China
| | - Miae Oh
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Jeonju-si 55365, Jeollabuk-do, Republic of Korea; (S.E.K.); (W.-S.S.); (M.O.); (S.L.); (J.-G.N.); (H.L.); (P.L.)
| | - Seunghoon Lee
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Jeonju-si 55365, Jeollabuk-do, Republic of Korea; (S.E.K.); (W.-S.S.); (M.O.); (S.L.); (J.-G.N.); (H.L.); (P.L.)
| | - Jin-Gu No
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Jeonju-si 55365, Jeollabuk-do, Republic of Korea; (S.E.K.); (W.-S.S.); (M.O.); (S.L.); (J.-G.N.); (H.L.); (P.L.)
| | - Haesun Lee
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Jeonju-si 55365, Jeollabuk-do, Republic of Korea; (S.E.K.); (W.-S.S.); (M.O.); (S.L.); (J.-G.N.); (H.L.); (P.L.)
| | - Poongyeon Lee
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Jeonju-si 55365, Jeollabuk-do, Republic of Korea; (S.E.K.); (W.-S.S.); (M.O.); (S.L.); (J.-G.N.); (H.L.); (P.L.)
| | - Keon Bong Oh
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Jeonju-si 55365, Jeollabuk-do, Republic of Korea; (S.E.K.); (W.-S.S.); (M.O.); (S.L.); (J.-G.N.); (H.L.); (P.L.)
| |
Collapse
|
13
|
Li Q, Yang Q, Guo P, Feng Y, Wang S, Guo J, Tang Z, Yu W, Liao J. Mitophagy contributes to zinc-induced ferroptosis in porcine testis cells. Food Chem Toxicol 2023; 179:113950. [PMID: 37481227 DOI: 10.1016/j.fct.2023.113950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 07/14/2023] [Accepted: 07/16/2023] [Indexed: 07/24/2023]
Abstract
Zinc (Zn) is a critical microelement for physiological process, but excess exposure can cause testicular dysfunction. However, the underlying mechanism of Zn-induced ferroptosis via regulating mitophagy is unknown. In this study, a total of 60 male weaned pigs were randomly divided into three groups and the content of Zn were 75 mg/kg (control), 750 mg/kg (Zn-I), 1500 mg/kg (Zn-II). Meanwhile, testicular cells were treated with ZnSO4 (0, 50 and 100 μM), and in combination of ZnSO4 (100 μM) and ferrostation-1, ML-210, or 3-methyladenine for 24 h. Our results verified that Zn could cause ferroptosis and lipid peroxidation, which were characterized by down-regulating level of SLC7A11, GPX4, and ferritin, and up-regulating levels of MDA, CD71, TF, and HMGB1 by Western blot, immunohistochemistry, immunofluorescence, peroxidase assay, et.ac. The opposite effect was shown after treatment with ferrostation-1 or ML-210. Meanwhile, the mitophagy-related proteins (PINK, Parkin, ATG5, LC3-II/LC3-I) were significantly upregulated in vivo and in vitro. Most importantly, 3-methyladenine observably relieved ferroptosis under Zn treatment through inhibiting mitophagy. Collectively, we demonstrated that mitophagy contributes to Zn-induced ferroptosis in porcine testis cells, providing a new insight into Zn toxicology.
Collapse
Affiliation(s)
- Quanwei Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, PR China
| | - Qingwen Yang
- Laboratory of Veterinary Pharmacology, Department of Animal Science and Technology, Chongqing Three Gorges Vocational College, Chongqing, PR China
| | - Pan Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, PR China
| | - Yuanhong Feng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, PR China
| | - Shaofeng Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, PR China
| | - Jianying Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, PR China
| | - Zhaoxin Tang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, PR China
| | - Wenlan Yu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, PR China.
| | - Jianzhao Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, Guangdong, PR China.
| |
Collapse
|
14
|
Tanihara F, Hirata M, Namula Z, Wittayarat M, Do LTK, Lin Q, Takebayashi K, Hara H, Nagahara M, Otoi T. GHR-mutant pig derived from domestic pig and microminipig hybrid zygotes using CRISPR/Cas9 system. Mol Biol Rep 2023; 50:5049-5057. [PMID: 37101010 DOI: 10.1007/s11033-023-08388-3] [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: 04/05/2022] [Accepted: 03/15/2023] [Indexed: 04/28/2023]
Abstract
BACKGROUND Pigs are excellent large animal models with several similarities to humans. They provide valuable insights into biomedical research that are otherwise difficult to obtain from rodent models. However, even if miniature pig strains are used, their large stature compared with other experimental animals requires a specific maintenance facility which greatly limits their usage as animal models. Deficiency of growth hormone receptor (GHR) function causes small stature phenotypes. The establishment of miniature pig strains via GHR modification will enhance their usage as animal models. Microminipig is an incredibly small miniature pig strain developed in Japan. In this study, we generated a GHR mutant pig using electroporation-mediated introduction of the CRISPR/Cas9 system into porcine zygotes derived from domestic porcine oocytes and microminipig spermatozoa. METHODS AND RESULTS First, we optimized the efficiency of five guide RNAs (gRNAs) designed to target GHR in zygotes. Embryos that had been electroporated with the optimized gRNAs and Cas9 were then transferred into recipient gilts. After embryo transfer, 10 piglets were delivered, and one carried a biallelic mutation in the GHR target region. The GHR biallelic mutant showed a remarkable growth-retardation phenotype. Furthermore, we obtained F1 pigs derived from the mating of GHR biallelic mutant with wild-type microminipig, and GHR biallelic mutant F2 pigs through sib-mating of F1 pigs. CONCLUSIONS We have successfully demonstrated the generation of biallelic GHR-mutant small-stature pigs. Backcrossing of GHR-deficient pig with microminipig will establish the smallest pig strain which can contribute significantly to the field of biomedical research.
Collapse
Affiliation(s)
- Fuminori Tanihara
- Faculty of Bioscience and Bioindustry, Tokushima University, Ishii, Myozai-gun, Tokushima, 7793233, Japan
| | - Maki Hirata
- Faculty of Bioscience and Bioindustry, Tokushima University, Ishii, Myozai-gun, Tokushima, 7793233, Japan
- Bio-Innovation Research Center, Tokushima University, Ishii, Myozai-gun, Tokushima, 7793233, Japan
| | - Zhao Namula
- Faculty of Bioscience and Bioindustry, Tokushima University, Ishii, Myozai-gun, Tokushima, 7793233, Japan
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Manita Wittayarat
- Faculty of Veterinary Science, Prince of Songkla University, Songkhla, 90110, Thailand
| | - Lanh Thi Kim Do
- Faculty of Bioscience and Bioindustry, Tokushima University, Ishii, Myozai-gun, Tokushima, 7793233, Japan
- Faculty of Veterinary Medicine, Vietnam National University of Agriculture, Hanoi, 100000, Vietnam
| | - Qingyi Lin
- Faculty of Bioscience and Bioindustry, Tokushima University, Ishii, Myozai-gun, Tokushima, 7793233, Japan
- Bio-Innovation Research Center, Tokushima University, Ishii, Myozai-gun, Tokushima, 7793233, Japan
| | - Koki Takebayashi
- Faculty of Bioscience and Bioindustry, Tokushima University, Ishii, Myozai-gun, Tokushima, 7793233, Japan
- Bio-Innovation Research Center, Tokushima University, Ishii, Myozai-gun, Tokushima, 7793233, Japan
| | - Hiromasa Hara
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, Tochigi, 3290498, Japan
- Laboratory of Regenerative And Cellular Medicine, Jichi Medical University, Tochigi, 3290498, Japan
| | - Megumi Nagahara
- Faculty of Bioscience and Bioindustry, Tokushima University, Ishii, Myozai-gun, Tokushima, 7793233, Japan
- Bio-Innovation Research Center, Tokushima University, Ishii, Myozai-gun, Tokushima, 7793233, Japan
| | - Takeshige Otoi
- Faculty of Bioscience and Bioindustry, Tokushima University, Ishii, Myozai-gun, Tokushima, 7793233, Japan.
- Bio-Innovation Research Center, Tokushima University, Ishii, Myozai-gun, Tokushima, 7793233, Japan.
| |
Collapse
|
15
|
Knoernschild K, Johnson HJ, Schroeder KE, Swier VJ, White KA, Sato TS, Rogers CS, Weimer JM, Sieren JC. Magnetic resonance brain volumetry biomarkers of CLN2 Batten disease identified with miniswine model. Sci Rep 2023; 13:5146. [PMID: 36991106 PMCID: PMC10060411 DOI: 10.1038/s41598-023-32071-z] [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: 09/29/2022] [Accepted: 03/22/2023] [Indexed: 03/31/2023] Open
Abstract
Late-infantile neuronal ceroid lipofuscinosis type 2 (CLN2) disease (Batten disease) is a rare pediatric disease, with symptom development leading to clinical diagnosis. Early diagnosis and effective tracking of disease progression are required for treatment. We hypothesize that brain volumetry is valuable in identifying CLN2 disease at an early stage and tracking disease progression in a genetically modified miniswine model. CLN2R208X/R208X miniswine and wild type controls were evaluated at 12- and 17-months of age, correlating to early and late stages of disease progression. Magnetic resonance imaging (MRI) T1- and T2-weighted data were acquired. Total intercranial, gray matter, cerebrospinal fluid, white matter, caudate, putamen, and ventricle volumes were calculated and expressed as proportions of the intracranial volume. The brain regions were compared between timepoints and cohorts using Gardner-Altman plots, mean differences, and confidence intervals. At an early stage of disease, the total intracranial volume (- 9.06 cm3), gray matter (- 4.37% 95 CI - 7.41; - 1.83), caudate (- 0.16%, 95 CI - 0.24; - 0.08) and putamen (- 0.11% 95 CI - 0.23; - 0.02) were all notably smaller in CLN2R208X/R208X miniswines versus WT, while cerebrospinal fluid was larger (+ 3.42%, 95 CI 2.54; 6.18). As the disease progressed to a later stage, the difference between the gray matter (- 8.27%, 95 CI - 10.1; - 5.56) and cerebrospinal fluid (+ 6.88%, 95 CI 4.31; 8.51) continued to become more pronounced, while others remained stable. MRI brain volumetry in this miniswine model of CLN2 disease is sensitive to early disease detection and longitudinal change monitoring, providing a valuable tool for pre-clinical treatment development and evaluation.
Collapse
Affiliation(s)
- Kevin Knoernschild
- Department of Radiology, University of Iowa, 200 Hawkins Drive cc704 GH, Iowa City, IA, 52242, USA
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA
| | - Hans J Johnson
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA
- Department of Electrical and Computer Engineering, University of Iowa, Iowa City, IA, USA
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Kimberly E Schroeder
- Department of Radiology, University of Iowa, 200 Hawkins Drive cc704 GH, Iowa City, IA, 52242, USA
| | - Vicki J Swier
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Katherine A White
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Takashi S Sato
- Department of Radiology, University of Iowa, 200 Hawkins Drive cc704 GH, Iowa City, IA, 52242, USA
| | | | - Jill M Weimer
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | - Jessica C Sieren
- Department of Radiology, University of Iowa, 200 Hawkins Drive cc704 GH, Iowa City, IA, 52242, USA.
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA.
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
16
|
Tardiolo G, Romeo O, Zumbo A, Di Marsico M, Sutera AM, Cigliano RA, Paytuví A, D’Alessandro E. Characterization of the Nero Siciliano Pig Fecal Microbiota after a Liquid Whey-Supplemented Diet. Animals (Basel) 2023; 13:642. [PMID: 36830429 PMCID: PMC9951753 DOI: 10.3390/ani13040642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/31/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
The utilization of dairy by-products as animal feed, especially in swine production, is a strategy to provide functional ingredients to improve gut health. This study explored the potential effect of a liquid whey-supplemented diet on the fecal microbiota of eleven pigs belonging to the Nero Siciliano breed. Five pigs were assigned to the control group and fed with a standard formulation feed, whereas six pigs were assigned to the experimental group and fed with the same feed supplemented with liquid whey. Fecal samples were collected from each individual before the experimental diet (T0), and one (T1) and two (T2) months after the beginning of the co-feed supplementation. Taxonomic analysis, based on the V3-V4 region of the bacterial 16S rRNA, showed that pig feces were populated by a complex microbial community with a remarkable abundance of Firmicutes, Bacteroidetes, and Spirochaetes phyla and Prevotella, Lactobacillus, Clostridium, and Treponema genera. Alpha and beta diversity values suggested that the experimental diet did not significantly affect the overall fecal microbiota diversity. However, analysis of abundance at different time points revealed significant variation in several bacterial genera, suggesting that the experimental diet potentially affected some genera of the microbial community.
Collapse
Affiliation(s)
- Giuseppe Tardiolo
- Department of Veterinary Sciences, University of Messina, Polo Universitario dell’Annunziata, Via Palatucci snc, 98168 Messina, Italy
| | - Orazio Romeo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Alessandro Zumbo
- Department of Veterinary Sciences, University of Messina, Polo Universitario dell’Annunziata, Via Palatucci snc, 98168 Messina, Italy
| | - Marco Di Marsico
- Sequentia Biotech SL, Carrer del Dr. Trueta 179, 08005 Barcelona, Spain
| | - Anna Maria Sutera
- Department of Veterinary Sciences, University of Messina, Polo Universitario dell’Annunziata, Via Palatucci snc, 98168 Messina, Italy
| | | | - Andreu Paytuví
- Sequentia Biotech SL, Carrer del Dr. Trueta 179, 08005 Barcelona, Spain
| | - Enrico D’Alessandro
- Department of Veterinary Sciences, University of Messina, Polo Universitario dell’Annunziata, Via Palatucci snc, 98168 Messina, Italy
| |
Collapse
|
17
|
Tanihara F, Hirata M, Otoi T. GEEP Method: An Optimized Electroporation-Mediated Gene Editing Approach for Establishment of Knockout Pig Lines. Methods Mol Biol 2023; 2637:293-300. [PMID: 36773155 DOI: 10.1007/978-1-0716-3016-7_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2023]
Abstract
Pigs are excellent large animal models owing to their several physiological and anatomical similarities to humans. Somatic cell nuclear transfer using gene-modified cells is the mainstream approach for generating genetically modified pigs. Recent advances in improving gene editors such as the CRISPR/Cas9 system have enabled direct gene modification in zygotes/embryos. Here, we describe the gene editing by electroporation of Cas9 protein (GEEP) method, an optimized electroporation-mediated method for the introduction of CRISPR/Cas9 into porcine zygotes/embryos. The simplicity and micromanipulation-free procedures are the major advantages of this method.
Collapse
Affiliation(s)
- Fuminori Tanihara
- Center for Development of Advanced Medical Technology, Jichi Medical University, Tochigi, Japan
| | - Maki Hirata
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
- Bio-Innovation Research Center, Tokushima University, Tokushima, Japan
| | - Takeshige Otoi
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan.
- Bio-Innovation Research Center, Tokushima University, Tokushima, Japan.
| |
Collapse
|
18
|
Yamashita MS, Melo EO. Animal Transgenesis and Cloning: Combined Development and Future Perspectives. Methods Mol Biol 2023; 2647:121-149. [PMID: 37041332 DOI: 10.1007/978-1-0716-3064-8_6] [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: 04/13/2023]
Abstract
The revolution in animal transgenesis began in 1981 and continues to become more efficient, cheaper, and faster to perform. New genome editing technologies, especially CRISPR-Cas9, are leading to a new era of genetically modified or edited organisms. Some researchers advocate this new era as the time of synthetic biology or re-engineering. Nonetheless, we are witnessing advances in high-throughput sequencing, artificial DNA synthesis, and design of artificial genomes at a fast pace. These advances in symbiosis with animal cloning by somatic cell nuclear transfer (SCNT) allow the development of improved livestock, animal models of human disease, and heterologous production of bioproducts for medical applications. In the context of genetic engineering, SCNT remains a useful technology to generate animals from genetically modified cells. This chapter addresses these fast-developing technologies driving this biotechnological revolution and their association with animal cloning technology.
Collapse
Affiliation(s)
- Melissa S Yamashita
- Embrapa Genetic Resources and Biotechnology, Brasília, Distrito Federal, Brazil
- Graduation Program in Animal Biology, University of Brasília, Brasília, Distrito Federal, Brazil
| | - Eduardo O Melo
- Embrapa Genetic Resources and Biotechnology, Brasília, Distrito Federal, Brazil.
- Graduation Program in Biotechnology, University of Tocantins, Gurupi, Tocantins, Brazil.
| |
Collapse
|
19
|
Wei J, Zhang W, Li J, Jin Y, Qiu Z. Application of the transgenic pig model in biomedical research: A review. Front Cell Dev Biol 2022; 10:1031812. [PMID: 36325365 PMCID: PMC9618879 DOI: 10.3389/fcell.2022.1031812] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 09/26/2022] [Indexed: 11/18/2022] Open
Abstract
The large animal model has gradually become an essential part of preclinical research studies, relating to exploring the disease pathological mechanism, genic function, pharmacy, and other subjects. Although the mouse model has already been widely accepted in clinical experiments, the need for finding an animal model with high similarity compared with a human model is urgent due to the different body functions and systems between mice and humans. The pig is an optimal choice for replacement. Therefore, enhancing the production of pigs used for models is an important part of the large animal model as well. Transgenic pigs show superiority in pig model creation because of the progress in genetic engineering. Successful cases of transgenic pig models occur in the clinical field of metabolic diseases, neurodegenerative diseases, and genetic diseases. In addition, the choice of pig breed influences the effort and efficiency of reproduction, and the mini pig has relative obvious advantages in pig model production. Indeed, pig models in these diseases provide great value in studies of their causes and treatments, especially at the genetic level. This review briefly outlines the method used to create transgenic pigs and species of producing transgenic pigs and provides an overview of their applications on different diseases and limitations for present pig model developments.
Collapse
Affiliation(s)
| | | | | | - Ye Jin
- *Correspondence: Ye Jin, ; Zhidong Qiu,
| | | |
Collapse
|
20
|
Wiarda JE, Trachsel JM, Sivasankaran SK, Tuggle CK, Loving CL. Intestinal single-cell atlas reveals novel lymphocytes in pigs with similarities to human cells. Life Sci Alliance 2022; 5:e202201442. [PMID: 35995567 PMCID: PMC9396248 DOI: 10.26508/lsa.202201442] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 08/04/2022] [Accepted: 08/04/2022] [Indexed: 12/13/2022] Open
Abstract
Lymphocytes can heavily influence intestinal health, but resolving intestinal lymphocyte function is challenging as the intestine contains a vastly heterogeneous mixture of cells. Pigs are an advantageous biomedical model, but deeper understanding of intestinal lymphocytes is warranted to improve model utility. Twenty-six cell types were identified in the porcine ileum by single-cell RNA sequencing and further compared with cells in human and murine ileum. Though general consensus of cell subsets across species was revealed, some porcine-specific lymphocyte subsets were identified. Differential tissue dissection and in situ analyses conferred spatial context, revealing similar locations of lymphocyte subsets in Peyer's patches and epithelium in pig-to-human comparisons. Like humans, activated and effector lymphocytes were abundant in the ileum but not periphery of pigs, suggesting tissue-specific and/or activation-associated gene expression. Gene signatures for peripheral and ileal innate lymphoid cells newly discovered in pigs were defined and highlighted similarities to human innate lymphoid cells. Overall, we reveal novel lymphocyte subsets in pigs and highlight utility of pigs for intestinal research applications.
Collapse
Affiliation(s)
- Jayne E Wiarda
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
- Immunobiology Graduate Program, Iowa State University, Ames, IA, USA
- Oak Ridge Institute for Science and Education, Agricultural Research Service Participation Program, Oak Ridge, TN, USA
| | - Julian M Trachsel
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
| | - Sathesh K Sivasankaran
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
- Genome Informatics Facility, Iowa State University, Ames, IA, USA
| | | | - Crystal L Loving
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, USA
| |
Collapse
|
21
|
Samiec M, Wiater J, Wartalski K, Skrzyszowska M, Trzcińska M, Lipiński D, Jura J, Smorąg Z, Słomski R, Duda M. The Relative Abundances of Human Leukocyte Antigen-E, α-Galactosidase A and α-Gal Antigenic Determinants Are Biased by Trichostatin A-Dependent Epigenetic Transformation of Triple-Transgenic Pig-Derived Dermal Fibroblast Cells. Int J Mol Sci 2022; 23:ijms231810296. [PMID: 36142211 PMCID: PMC9499218 DOI: 10.3390/ijms231810296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/01/2022] [Accepted: 09/04/2022] [Indexed: 11/16/2022] Open
Abstract
The present study sought to establish the mitotically stable adult cutaneous fibroblast cell (ACFC) lines stemming from hFUT2×hGLA×HLA-E triple-transgenic pigs followed by trichostatin A (TSA)-assisted epigenetically modulating the reprogrammability of the transgenes permanently incorporated into the host genome and subsequent comprehensive analysis of molecular signatures related to proteomically profiling the generated ACFC lines. The results of Western blot and immunofluorescence analyses have proved that the profiles of relative abundance (RA) noticed for both recombinant human α-galactosidase A (rhα-Gal A) and human leukocyte antigen-E (HLA-E) underwent significant upregulations in tri-transgenic (3×TG) ACFCs subjected to TSA-mediated epigenetic transformation as compared to not only their TSA-unexposed counterparts but also TSA-treated and untreated non-transgenic (nTG) cells. The RT-qPCR-based analysis of porcine tri-genetically engineered ACFCs revealed stable expression of mRNA fractions transcribed from hFUT2, hGLA and HLA-E transgenes as compared to a lack of such transcriptional activities in non-transgenic ACFC variants. Furthermore, although TSA-based epigenomic modulation has given rise to a remarkable increase in the expression levels of Galα1→3Gal (α-Gal) epitopes that have been determined by lectin blotting analysis, their semi-quantitative profiles have dwindled profoundly in both TSA-exposed and unexposed 3×TG ACFCs as compared to their nTG counterparts. In conclusion, thoroughly exploring proteomic signatures in such epigenetically modulated ex vivo models devised on hFUT2×hGLA×HLA-E triple-transgenic ACFCs that display augmented reprogrammability of translational activities of two mRNA transcripts coding for rhα-Gal A and HLA-E proteins might provide a completely novel and powerful research tool for the panel of further studies. The objective of these future studies should be to multiply the tri-transgenic pigs with the aid of somatic cell nuclear transfer (SCNT)-based cloning for the purposes of both xenografting the porcine cutaneous bioprostheses and dermoplasty-mediated surgical treatments in human patients.
Collapse
Affiliation(s)
- Marcin Samiec
- Department of Reproductive Biotechnology and Cryoconservation, National Research Institute of Animal Production, Krakowska 1 Street, 32-083 Balice, Poland
- Correspondence: (M.S.); (J.W.)
| | - Jerzy Wiater
- Department of Histology, Jagiellonian University Medical College, Kopernika 7 Street, 31-034 Kraków, Poland
- Correspondence: (M.S.); (J.W.)
| | - Kamil Wartalski
- Department of Histology, Jagiellonian University Medical College, Kopernika 7 Street, 31-034 Kraków, Poland
| | - Maria Skrzyszowska
- Department of Reproductive Biotechnology and Cryoconservation, National Research Institute of Animal Production, Krakowska 1 Street, 32-083 Balice, Poland
| | - Monika Trzcińska
- Department of Reproductive Biotechnology and Cryoconservation, National Research Institute of Animal Production, Krakowska 1 Street, 32-083 Balice, Poland
| | - Daniel Lipiński
- Department of Biochemistry and Biotechnology, Poznań University of Life Sciences, Dojazd 11 Street, 60-647 Poznań, Poland
| | - Jacek Jura
- Department of Reproductive Biotechnology and Cryoconservation, National Research Institute of Animal Production, Krakowska 1 Street, 32-083 Balice, Poland
| | - Zdzisław Smorąg
- Department of Reproductive Biotechnology and Cryoconservation, National Research Institute of Animal Production, Krakowska 1 Street, 32-083 Balice, Poland
| | - Ryszard Słomski
- Department of Biochemistry and Biotechnology, Poznań University of Life Sciences, Dojazd 11 Street, 60-647 Poznań, Poland
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32 Street, 60-479 Poznań, Poland
| | - Małgorzata Duda
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University in Krakow, Gronostajowa 9 Street, 30-387 Kraków, Poland
| |
Collapse
|
22
|
Abstract
The genetic modification of pigs as a source of transplantable organs is one of several possible solutions to the chronic organ shortage. This paper describes existing ethical tensions in xenotransplantation (XTx) that argue against pursuing it. Recommendations for lifelong infectious disease surveillance and notification of close contacts of recipients are in tension with the rights of human research subjects. Parental/guardian consent for pediatric xenograft recipients is in tension with a child's right to an open future. Individual consent to transplant is in tension with public health threats that include zoonotic diseases. XTx amplifies concerns about justice in organ transplantation and could exacerbate existing inequities. The prevention of infectious disease in source animals is in tension with the best practices of animal care and animal welfare, requiring isolation, ethologically inappropriate housing, and invasive reproductive procedures that would severely impact the well-being of intelligent social creatures like pigs.
Collapse
|
23
|
Koehler N, Buhler L, Egger B, Gonelle-Gispert C. Multipotent Mesenchymal Stromal Cells Interact and Support Islet of Langerhans Viability and Function. Front Endocrinol (Lausanne) 2022; 13:822191. [PMID: 35222280 PMCID: PMC8864309 DOI: 10.3389/fendo.2022.822191] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/13/2022] [Indexed: 11/13/2022] Open
Abstract
Type 1 diabetes (T1D) is a widespread disease, affecting approximately 41.5 million people worldwide. It is generally treated with exogenous insulin, maintaining physiological blood glucose levels but also leading to long-term therapeutic complications. Pancreatic islet cell transplantation offers a potential alternative treatment to insulin injections. Shortage of human organ donors has raised the interest for porcine islet xenotransplantation. Neonatal porcine islets are highly available, can proliferate and mature in vitro as well as after transplantation in vivo. Despite promising preclinical results, delayed insulin secretion caused by immaturity and immunogenicity of the neonatal porcine islets remains a challenge for their clinical application. Multipotent mesenchymal stromal cells (MSCs) are known to have pro-angiogenic, anti-inflammatory and immunomodulatory effects. The current state of research emphasizes the great potential of co-culture and co-transplantation of islet cells with MSCs. Studies have shown enhanced islet proliferation and maturation, insulin secretion and graft survival, resulting in an improved graft outcome. This review summarizes the immunomodulatory and anti-inflammatory properties of MSC in the context of islet transplantation.
Collapse
Affiliation(s)
- Naomi Koehler
- Surgical Research Unit, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Leo Buhler
- Department of Surgery, Cantonal Hospital Fribourg, Fribourg, Switzerland
| | - Bernhard Egger
- Surgical Research Unit, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
- Department of Surgery, Cantonal Hospital Fribourg, Fribourg, Switzerland
| | - Carmen Gonelle-Gispert
- Surgical Research Unit, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
- *Correspondence: Carmen Gonelle-Gispert,
| |
Collapse
|
24
|
Hirata M, Wittayarat M, Namula Z, Le QA, Lin Q, Takebayashi K, Thongkittidilok C, Mito T, Tomonari S, Tanihara F, Otoi T. Generation of mutant pigs by lipofection-mediated genome editing in embryos. Sci Rep 2021; 11:23806. [PMID: 34903813 PMCID: PMC8668999 DOI: 10.1038/s41598-021-03325-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 11/30/2021] [Indexed: 01/03/2023] Open
Abstract
The specificity and efficiency of CRISPR/Cas9 gene-editing systems are determined by several factors, including the mode of delivery, when applied to mammalian embryos. Given the limited time window for delivery, faster and more reliable methods to introduce Cas9-gRNA ribonucleoprotein complexes (RNPs) into target embryos are needed. In pigs, somatic cell nuclear transfer using gene-modified somatic cells and the direct introduction of gene editors into the cytoplasm of zygotes/embryos by microinjection or electroporation have been used to generate gene-edited embryos; however, these strategies require expensive equipment and sophisticated techniques. In this study, we developed a novel lipofection-mediated RNP transfection technique that does not require specialized equipment for the generation of gene-edited pigs and produced no detectable off-target events. In particular, we determined the concentration of lipofection reagent for efficient RNP delivery into embryos and successfully generated MSTN gene-edited pigs (with mutations in 7 of 9 piglets) after blastocyst transfer to a recipient gilt. This newly established lipofection-based technique is still in its early stages and requires improvements, particularly in terms of editing efficiency. Nonetheless, this practical method for rapid and large-scale lipofection-mediated gene editing in pigs has important agricultural and biomedical applications.
Collapse
Affiliation(s)
- Maki Hirata
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan.,Bio-Innovation Research Center, Tokushima University, Tokushima, Japan
| | - Manita Wittayarat
- Faculty of Veterinary Science, Prince of Songkla University, Songkhla, Thailand
| | - Zhao Namula
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan.,College of Coastal Agricultural Sciences, Guangdong Ocean University, Guangdong, China
| | - Quynh Anh Le
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - Qingyi Lin
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - Koki Takebayashi
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | | | - Taro Mito
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan.,Bio-Innovation Research Center, Tokushima University, Tokushima, Japan
| | - Sayuri Tomonari
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - Fuminori Tanihara
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan. .,Center for Development of Advanced Medical Technology, Jichi Medical University, Tochigi, Japan.
| | - Takeshige Otoi
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan.,Bio-Innovation Research Center, Tokushima University, Tokushima, Japan
| |
Collapse
|
25
|
A Dystrophin Exon-52 Deleted Miniature Pig Model of Duchenne Muscular Dystrophy and Evaluation of Exon Skipping. Int J Mol Sci 2021; 22:ijms222313065. [PMID: 34884867 PMCID: PMC8657897 DOI: 10.3390/ijms222313065] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/24/2021] [Accepted: 11/25/2021] [Indexed: 12/12/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal X-linked recessive disorder caused by mutations in the DMD gene and the subsequent lack of dystrophin protein. Recently, phosphorodiamidate morpholino oligomer (PMO)-antisense oligonucleotides (ASOs) targeting exon 51 or 53 to reestablish the DMD reading frame have received regulatory approval as commercially available drugs. However, their applicability and efficacy remain limited to particular patients. Large animal models and exon skipping evaluation are essential to facilitate ASO development together with a deeper understanding of dystrophinopathies. Using recombinant adeno-associated virus-mediated gene targeting and somatic cell nuclear transfer, we generated a Yucatan miniature pig model of DMD with an exon 52 deletion mutation equivalent to one of the most common mutations seen in patients. Exon 52-deleted mRNA expression and dystrophin deficiency were confirmed in the skeletal and cardiac muscles of DMD pigs. Accordingly, dystrophin-associated proteins failed to be recruited to the sarcolemma. The DMD pigs manifested early disease onset with severe bodywide skeletal muscle degeneration and with poor growth accompanied by a physical abnormality, but with no obvious cardiac phenotype. We also demonstrated that in primary DMD pig skeletal muscle cells, the genetically engineered exon-52 deleted pig DMD gene enables the evaluation of exon 51 or 53 skipping with PMO and its advanced technology, peptide-conjugated PMO. The results show that the DMD pigs developed here can be an appropriate large animal model for evaluating in vivo exon skipping efficacy.
Collapse
|
26
|
Gao Q, Davis R, Fitch Z, Mulvihill M, Ezekian B, Schroder P, Schmitz R, Song M, Leopardi F, Ribeiro M, Miller A, Moris D, Shaw B, Samy K, Reimann K, Williams K, Collins B, Kirk AD. Anti-thymoglobulin induction improves neonatal porcine xenoislet engraftment and survival. Xenotransplantation 2021; 28:e12713. [PMID: 34951057 PMCID: PMC8715890 DOI: 10.1111/xen.12713] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/13/2021] [Accepted: 09/07/2021] [Indexed: 12/18/2022]
Abstract
Porcine islet xenotransplantation is a viable strategy to treat diabetes. Its translation has been limited by the pre-clinical development of a clinically available immunosuppressive regimen. We tested two clinically relevant induction agents in a non-human primate (NHP) islet xenotransplantation model to compare depletional versus nondepletional induction immunosuppression. Neonatal porcine islets were isolated from GKO or hCD46/GKO transgenic piglets and transplanted via portal vein infusion in diabetic rhesus macaques. Induction therapy consisted of either basiliximab (n = 6) or rhesus-specific anti-thymocyte globulin (rhATG, n = 6), combined with a maintenance regimen using B7 costimulation blockade, tacrolimus with a delayed transition to sirolimus, and mycophenolate mofetil. Xenografts were monitored by blood glucose levels and porcine C-peptide measurements. Of the six receiving basiliximab induction, engraftment was achieved in 4 with median graft survival of 14 days. All six receiving rhATG induction engrafted with significantly longer xenograft survival at 40.5 days (P = 0.03). These data suggest that depletional induction provides superior xenograft survival to nondepletional induction, in the setting of a costimulation blockade-based maintenance regimen.
Collapse
Affiliation(s)
- Qimeng Gao
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, 27710, USA
| | - Robert Davis
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, 27710, USA
| | - Zachary Fitch
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, 27710, USA
| | - Michael Mulvihill
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, 27710, USA
| | - Brian Ezekian
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, 27710, USA
| | - Paul Schroder
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, 27710, USA
| | - Robin Schmitz
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, 27710, USA
| | - Mingqing Song
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, 27710, USA
| | - Frank Leopardi
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, 27710, USA
| | - Marianna Ribeiro
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, 27710, USA
| | - Allison Miller
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, 27710, USA
| | - Dimitrios Moris
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, 27710, USA
| | - Brian Shaw
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, 27710, USA
| | - Kannan Samy
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, 27710, USA
| | - Keith Reimann
- MassBiologics, University of Massachusetts Medical School, Worcester, Massachusetts, 01655, USA
| | - Kyha Williams
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, 27710, USA
| | - Bradley Collins
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, 27710, USA
| | - Allan D Kirk
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, 27710, USA
| |
Collapse
|
27
|
Jeong PS, Yang HJ, Park SH, Gwon MA, Joo YE, Kim MJ, Kang HG, Lee S, Park YH, Song BS, Kim SU, Koo DB, Sim BW. Combined Chaetocin/Trichostatin A Treatment Improves the Epigenetic Modification and Developmental Competence of Porcine Somatic Cell Nuclear Transfer Embryos. Front Cell Dev Biol 2021; 9:709574. [PMID: 34692674 PMCID: PMC8526721 DOI: 10.3389/fcell.2021.709574] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 09/15/2021] [Indexed: 01/03/2023] Open
Abstract
Developmental defects in somatic cell nuclear transfer (SCNT) embryos are principally attributable to incomplete epigenetic reprogramming. Small-molecule inhibitors such as histone methyltransferase inhibitors (HMTi) and histone deacetylase inhibitors (HDACi) have been used to improve reprogramming efficiency of SCNT embryos. However, their possible synergistic effect on epigenetic reprogramming has not been studied. In this study, we explored whether combined treatment with an HMTi (chaetocin) and an HDACi (trichostatin A; TSA) synergistically enhanced epigenetic reprogramming and the developmental competence of porcine SCNT embryos. Chaetocin, TSA, and the combination significantly increased the cleavage and blastocyst formation rate, hatching/hatched blastocyst rate, and cell numbers and survival rate compared to control embryos. In particular, the combined treatment improved the rate of development to blastocysts more so than chaetocin or TSA alone. TSA and combined chaetocin/TSA significantly reduced the H3K9me3 levels and increased the H3K9ac levels in SCNT embryos, although chaetocin alone significantly reduced only the H3K9me3 levels. Moreover, these inhibitors also decreased global DNA methylation in SCNT embryos. In addition, the expression of zygotic genome activation- and imprinting-related genes was increased by chaetocin or TSA, and more so by the combination, to levels similar to those of in vitro-fertilized embryos. These results suggest that combined chaetocin/TSA have synergistic effects on improving the developmental competences by regulating epigenetic reprogramming and correcting developmental potential-related gene expression in porcine SCNT embryos. Therefore, these strategies may contribute to the generation of transgenic pigs for biomedical research.
Collapse
Affiliation(s)
- Pil-Soo Jeong
- Futuristic Animal Resource and Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, South Korea.,Department of Biotechnology, College of Engineering, Daegu University, Gyeongsan, South Korea
| | - Hae-Jun Yang
- Futuristic Animal Resource and Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, South Korea
| | - Soo-Hyun Park
- Futuristic Animal Resource and Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, South Korea.,Department of Animal Science, College of Natural Resources and Life Science, Pusan National University, Miryang, South Korea
| | - Min Ah Gwon
- Futuristic Animal Resource and Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, South Korea.,Department of Biotechnology, College of Engineering, Daegu University, Gyeongsan, South Korea
| | - Ye Eun Joo
- Futuristic Animal Resource and Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, South Korea.,Department of Animal Science, College of Natural Resources and Life Science, Pusan National University, Miryang, South Korea
| | - Min Ju Kim
- Futuristic Animal Resource and Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, South Korea.,Department of Animal Science, College of Natural Resources and Life Science, Pusan National University, Miryang, South Korea
| | - Hyo-Gu Kang
- Futuristic Animal Resource and Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, South Korea.,Department of Animal Science and Biotechnology, College of Agriculture and Life Science, Chungnam National University, Daejeon, South Korea
| | - Sanghoon Lee
- Futuristic Animal Resource and Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, South Korea
| | - Young-Ho Park
- Futuristic Animal Resource and Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, South Korea
| | - Bong-Seok Song
- Futuristic Animal Resource and Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, South Korea
| | - Sun-Uk Kim
- Futuristic Animal Resource and Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, South Korea.,Department of Functional Genomics, University of Science and Technology, Daejeon, South Korea
| | - Deog-Bon Koo
- Department of Biotechnology, College of Engineering, Daegu University, Gyeongsan, South Korea
| | - Bo-Woong Sim
- Futuristic Animal Resource and Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, South Korea
| |
Collapse
|
28
|
Jiao D, Cheng W, Zhang X, Zhang Y, Guo J, Li Z, Shi D, Xiong Z, Qing Y, Jamal MA, Xu K, Zhao HY, Wei HJ. Improving porcine SCNT efficiency by selecting donor cells size. Cell Cycle 2021; 20:2264-2277. [PMID: 34583621 DOI: 10.1080/15384101.2021.1980983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Considerable advancements have recently been achieved in porcine somatic cell nuclear transfer (SCNT), but the efficiency remains low. Donor cell size might play an important role in SCNT, but its effects in pigs remain unclear. This study aimed to evaluate the efficiency of porcine SCNT by selecting donor cells of suitable size. Porcine fetal fibroblasts (PFFs) were divided into three groups, group S (small, d ≤ 13 μm), group M (medium, 13 μm<d ≤ 18 μm), and group L (large, d > 18 μm), and their biological characteristics were analyzed. Next, SCNT was performed using PFFs of different sizes to evaluate the developmental potential of reconstructed embryos. The data showed that PFFs in groups S, M and L accounted for 17.5%, 47.7% and 34.8% of cells, respectively. Morphologically, cells in group S exhibited clear and regular cell membranes and nuclei, whereas cells in groups M and L displayed varying degrees of cell membrane protuberance, karyo-pyknosis, autophagy and mitochondrial abnormalities. In addition, the growth status and proliferation capabilities of cells in group S were significantly better than those of group M and group L. The percentage of cells at G0/G1 in group S and M were significantly greater than group L. The senescence rate of group S was lower than group M and group L. The apoptosis rate of group S was significantly lower than that of group L but comparable to that of group M . The cleavage rate of group S was also significantly greater than that of group M but comparable to that of group L . The blastocyst rate of group S was significantly greater than that of group M and group L. The blastocyst cell numbers of group S were also significantly greater than those of group M and group L. These findings suggested that small PFFs with a diameter of less than 13 μm are more suitable donor cells for SCNT in pigs.Abbreviations: DMEM: Dulbecco's modified Eagle's medium; FBS: Fetal bovine serum; PBS: Phosphate buffer saline; PFFs: Porcine fetal fibroblast cells; SCNT: Somatic cell nuclear transfer.
Collapse
Affiliation(s)
- Deling Jiao
- Key laboratory for porcine gene editing and xenotransplantation in Yunnan Province, Yunnan Agricultural University, Kunming, China.,Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming, China.,College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Wenmin Cheng
- Key laboratory for porcine gene editing and xenotransplantation in Yunnan Province, Yunnan Agricultural University, Kunming, China.,Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming, China.,College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Xiaolin Zhang
- Key laboratory for porcine gene editing and xenotransplantation in Yunnan Province, Yunnan Agricultural University, Kunming, China.,Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming, China.,College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Yifan Zhang
- Key laboratory for porcine gene editing and xenotransplantation in Yunnan Province, Yunnan Agricultural University, Kunming, China.,Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming, China.,College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Jianxiong Guo
- Key laboratory for porcine gene editing and xenotransplantation in Yunnan Province, Yunnan Agricultural University, Kunming, China.,Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming, China.,College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Zhuo Li
- Key laboratory for porcine gene editing and xenotransplantation in Yunnan Province, Yunnan Agricultural University, Kunming, China.,Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming, China.,College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Dejia Shi
- Key laboratory for porcine gene editing and xenotransplantation in Yunnan Province, Yunnan Agricultural University, Kunming, China.,Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming, China.,College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Zhe Xiong
- Key laboratory for porcine gene editing and xenotransplantation in Yunnan Province, Yunnan Agricultural University, Kunming, China.,Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming, China.,College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Yubo Qing
- Key laboratory for porcine gene editing and xenotransplantation in Yunnan Province, Yunnan Agricultural University, Kunming, China.,Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming, China.,College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China.,College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Muhammad Ameen Jamal
- Key laboratory for porcine gene editing and xenotransplantation in Yunnan Province, Yunnan Agricultural University, Kunming, China.,Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming, China.,College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Kaixiang Xu
- Key laboratory for porcine gene editing and xenotransplantation in Yunnan Province, Yunnan Agricultural University, Kunming, China.,Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming, China.,College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Hong-Ye Zhao
- Key laboratory for porcine gene editing and xenotransplantation in Yunnan Province, Yunnan Agricultural University, Kunming, China.,Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming, China.,State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
| | - Hong-Jiang Wei
- Key laboratory for porcine gene editing and xenotransplantation in Yunnan Province, Yunnan Agricultural University, Kunming, China.,Xenotransplantation Research Engineering Center in Yunnan Province, Yunnan Agricultural University, Kunming, China.,College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China.,State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, China
| |
Collapse
|
29
|
Banana Lectin from Musa paradisiaca Is Mitogenic for Cow and Pig PBMC via IL-2 Pathway and ELF1. IMMUNO 2021. [DOI: 10.3390/immuno1030018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The aim of the study was to gain deeper insights in the potential of polyclonal stimulation of PBMC with banana lectin (BanLec) from Musa paradisiaca. BanLec induced a marked proliferative response in cow and pig PBMC, but was strongest in pigs, where it induced an even higher proliferation rate than Concanavalin A. Molecular processes associated with respective responses in porcine PBMC were examined with differential proteome analyses. Discovery proteomic experiments was applied to BanLec stimulated PBMC and cellular and secretome responses were analyzed with label free LC-MS/MS. In PBMC, 3955 proteins were identified. After polyclonal stimulation with BanLec, 459 proteins showed significantly changed abundance in PBMC. In respective PBMC secretomes, 2867 proteins were identified with 231 differentially expressed candidates as reaction to BanLec stimulation. The transcription factor “E74 like ETS transcription factor 1 (ELF1)” was solely enriched in BanLec stimulated PBMC. BanLec induced secretion of several immune regulators, amongst them positive regulators of activated T cell proliferation and Jak-STAT signaling pathway. Top changed immune proteins were CD226, CD27, IFNG, IL18, IL2, CXCL10, LAT, ICOS, IL2RA, LAG3, and CD300C. BanLec stimulates PBMC of cows and pigs polyclonally and induces IL2 pathway and further proinflammatory cytokines. Proteomics data are available via ProteomeXchange with identifier PXD027505.
Collapse
|
30
|
Giuffrè L, Giosa D, Galeano G, Aiese Cigliano R, Paytuví-Gallart A, Sutera AM, Tardiolo G, Zumbo A, Romeo O, D’Alessandro E. Whole-metagenome shotgun sequencing of pig faecal microbiome. ITALIAN JOURNAL OF ANIMAL SCIENCE 2021. [DOI: 10.1080/1828051x.2021.1952910] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Letterio Giuffrè
- Dipartimento di Scienze Veterinarie, Università di Messina, Messina, Italy
| | - Domenico Giosa
- Dipartimento di Medicina Clinica e Sperimentale, Università di Messina, Messina, Italy
| | - Grazia Galeano
- Dipartimento di Scienze Veterinarie, Università di Messina, Messina, Italy
| | | | | | - Anna Maria Sutera
- Dipartimento di Scienze Veterinarie, Università di Messina, Messina, Italy
| | - Giuseppe Tardiolo
- Dipartimento di Scienze Veterinarie, Università di Messina, Messina, Italy
| | - Alessandro Zumbo
- Dipartimento di Scienze Veterinarie, Università di Messina, Messina, Italy
| | - Orazio Romeo
- Dipartimento di Scienze Chimiche, Farmaceutiche ed Ambientali, Università di Messina, Biologiche, Messina, Italy
| | | |
Collapse
|
31
|
CRISPR/Cas Technology in Pig-to-Human Xenotransplantation Research. Int J Mol Sci 2021; 22:ijms22063196. [PMID: 33801123 PMCID: PMC8004187 DOI: 10.3390/ijms22063196] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 02/06/2023] Open
Abstract
CRISPR/Cas (clustered regularly interspaced short palindromic repeats linked to Cas nuclease) technology has revolutionized many aspects of genetic engineering research. Thanks to it, it became possible to study the functions and mechanisms of biology with greater precision, as well as to obtain genetically modified organisms, both prokaryotic and eukaryotic. The changes introduced by the CRISPR/Cas system are based on the repair paths of the single or double strand DNA breaks that cause insertions, deletions, or precise integrations of donor DNA. These changes are crucial for many fields of science, one of which is the use of animals (pigs) as a reservoir of tissues and organs for xenotransplantation into humans. Non-genetically modified animals cannot be used to save human life and health due to acute immunological reactions resulting from the phylogenetic distance of these two species. This review is intended to collect and summarize the advantages as well as achievements of the CRISPR/Cas system in pig-to-human xenotransplantation research. In addition, it demonstrates barriers and limitations that require careful evaluation before attempting to experiment with this technology.
Collapse
|
32
|
Interneuron Origins in the Embryonic Porcine Medial Ganglionic Eminence. J Neurosci 2021; 41:3105-3119. [PMID: 33637558 DOI: 10.1523/jneurosci.2738-20.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/18/2020] [Accepted: 01/08/2021] [Indexed: 02/06/2023] Open
Abstract
Interneurons contribute to the complexity of neural circuits and maintenance of normal brain function. Rodent interneurons originate in embryonic ganglionic eminences, but developmental origins in other species are less understood. Here, we show that transcription factor expression patterns in porcine embryonic subpallium are similar to rodents, delineating a distinct medial ganglionic eminence (MGE) progenitor domain. On the basis of Nkx2.1, Lhx6, and Dlx2 expression, in vitro differentiation into neurons expressing GABA, and robust migratory capacity in explant assays, we propose that cortical and hippocampal interneurons originate from a porcine MGE region. Following xenotransplantation into adult male and female rat hippocampus, we further demonstrate that porcine MGE progenitors, like those from rodents, migrate and differentiate into morphologically distinct interneurons expressing GABA. Our findings reveal that basic rules for interneuron development are conserved across species, and that porcine embryonic MGE progenitors could serve as a valuable source for interneuron-based xenotransplantation therapies.SIGNIFICANCE STATEMENT Here we demonstrate that porcine medial ganglionic eminence, like rodents, exhibit a distinct transcriptional and interneuron-specific antibody profile, in vitro migratory capacity and are amenable to xenotransplantation. This is the first comprehensive examination of embryonic interneuron origins in the pig; and because a rich neurodevelopmental literature on embryonic mouse medial ganglionic eminence exists (with some additional characterizations in other species, e.g., monkey and human), our work allows direct neurodevelopmental comparisons with this literature.
Collapse
|
33
|
Elkhadragy L, Regan MR, M Totura W, Goli KD, Patel S, Garcia K, Stewart M, Schook LB, Gaba RC, Schachtschneider KM. Generation of genetically tailored porcine liver cancer cells by CRISPR/Cas9 editing. Biotechniques 2021; 70:37-48. [PMID: 33222517 PMCID: PMC7852845 DOI: 10.2144/btn-2020-0119] [Citation(s) in RCA: 5] [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: 07/30/2020] [Accepted: 10/01/2020] [Indexed: 12/28/2022] Open
Abstract
Pigs provide a valuable large animal model for several diseases due to their similarity with humans in anatomy, physiology, genetics and drug metabolism. We recently generated a porcine model for TP53R167H and KRASG12D driven hepatocellular carcinoma (HCC) by autologous liver implantation. Here we describe a streamlined approach for developing genetically tailored porcine HCC cells by CRISPR/Cas9 gene editing and isolation of homogenous genetically validated cell clones. The combination of CRISPR/Cas9 editing of HCC cells described herein with the orthotopic HCC model enables development of various porcine HCC models, each with a specific mutational profile. This allows modeling the effect of different driver mutation combinations on tumor progression and in vivo testing of novel targeted therapeutic approaches in a clinically relevant large animal model.
Collapse
Affiliation(s)
- Lobna Elkhadragy
- Department of Radiology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Maureen R Regan
- Department of Biochemistry & Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - William M Totura
- Department of Radiology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Kimia Dasteh Goli
- Department of Radiology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Shovik Patel
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Kelly Garcia
- Biological Research Laboratory, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Matthew Stewart
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
| | - Lawrence B Schook
- Department of Radiology, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Ron C Gaba
- Department of Radiology, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Pathology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Kyle M Schachtschneider
- Department of Radiology, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Biochemistry & Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
- National Center for Supercomputing Applications, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|