1
|
Navarro M, Laiz-Quiroga L, Blüguermann C, Mutto A. Livestock embryonic stem cells for reproductive biotechniques and genetic improvement. Anim Reprod 2024; 21:e20240029. [PMID: 39175999 PMCID: PMC11340801 DOI: 10.1590/1984-3143-ar2024-0029] [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: 03/21/2024] [Accepted: 05/27/2024] [Indexed: 08/24/2024] Open
Abstract
Embryonic stem cells (ESCs) have proven to be a great in vitro model that faithfully recapitulates the events that occur during in vivo embryogenesis, making them a unique tool to study the cellular and molecular mechanisms that define tissue specification during embryonic development. Livestock ESCs are particularly attractive and have broad prospects including drug selection and human disease modeling, improvement of reproductive biotechniques and agriculture-related applications such as production of genetically modified animals. While mice and human ESCs have been established many years ago, no significant advances were made in livestock species until recently. Nowadays, livestock ESCs are available from cattle, pigs, sheep, horses and rabbits with different states of pluripotency. In this review, we summarize the current advances on livestock ESCs establishment and maintenance along with their present and future applications.
Collapse
Affiliation(s)
- Micaela Navarro
- Laboratorio de Biotecnologías aplicadas a la Reproducción Animal, Instituto de Investigaciones Biotecnológicas “Dr. Rodolfo Ugalde”, Universidad Nacional de General San Martín, Buenos Aires, Argentina
| | - Lucia Laiz-Quiroga
- Laboratorio de Biotecnologías aplicadas a la Reproducción Animal, Instituto de Investigaciones Biotecnológicas “Dr. Rodolfo Ugalde”, Universidad Nacional de General San Martín, Buenos Aires, Argentina
| | - Carolina Blüguermann
- Laboratorio de Biotecnologías aplicadas a la Reproducción Animal, Instituto de Investigaciones Biotecnológicas “Dr. Rodolfo Ugalde”, Universidad Nacional de General San Martín, Buenos Aires, Argentina
| | - Adrián Mutto
- Laboratorio de Biotecnologías aplicadas a la Reproducción Animal, Instituto de Investigaciones Biotecnológicas “Dr. Rodolfo Ugalde”, Universidad Nacional de General San Martín, Buenos Aires, Argentina
| |
Collapse
|
2
|
Najafi P, Reimer C, Gilthorpe JD, Jacobsen KR, Ramløse M, Paul NF, Simianer H, Tetens J, Falker-Gieske C. Genomic evidence for the suitability of Göttingen Minipigs with a rare seizure phenotype as a model for human epilepsy. Neurogenetics 2024; 25:103-117. [PMID: 38383918 PMCID: PMC11076379 DOI: 10.1007/s10048-024-00750-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/02/2024] [Indexed: 02/23/2024]
Abstract
Epilepsy is a complex genetic disorder that affects about 2% of the global population. Although the frequency and severity of epileptic seizures can be reduced by a range of pharmacological interventions, there are no disease-modifying treatments for epilepsy. The development of new and more effective drugs is hindered by a lack of suitable animal models. Available rodent models may not recapitulate all key aspects of the disease. Spontaneous epileptic convulsions were observed in few Göttingen Minipigs (GMPs), which may provide a valuable alternative animal model for the characterisation of epilepsy-type diseases and for testing new treatments. We have characterised affected GMPs at the genome level and have taken advantage of primary fibroblast cultures to validate the functional impact of fixed genetic variants on the transcriptome level. We found numerous genes connected to calcium metabolism that have not been associated with epilepsy before, such as ADORA2B, CAMK1D, ITPKB, MCOLN2, MYLK, NFATC3, PDGFD, and PHKB. Our results have identified two transcription factor genes, EGR3 and HOXB6, as potential key regulators of CACNA1H, which was previously linked to epilepsy-type disorders in humans. Our findings provide the first set of conclusive results to support the use of affected subsets of GMPs as an alternative and more reliable model system to study human epilepsy. Further neurological and pharmacological validation of the suitability of GMPs as an epilepsy model is therefore warranted.
Collapse
Affiliation(s)
- Pardis Najafi
- Department of Animal Sciences, Georg-August-University, Burckhardtweg 2, 37077, Göttingen, Germany
- Center for Integrated Breeding Research, Georg-August-University, Albrecht-Thaer-Weg 3, 37075, Göttingen, Germany
| | - Christian Reimer
- Center for Integrated Breeding Research, Georg-August-University, Albrecht-Thaer-Weg 3, 37075, Göttingen, Germany
- Friedrich-Loeffler-Institute, Federal Research Institute for Animal Health, Höltystr. 10, 31535, Neustadt, Germany
| | - Jonathan D Gilthorpe
- Department of Integrative Medical Biology, Umeå University, 901 87, Umeå, Sweden
| | - Kirsten R Jacobsen
- Ellegaard Göttingen Minipigs A/S, Sorø Landevej 302, 4261, Dalmose, Denmark
| | - Maja Ramløse
- Ellegaard Göttingen Minipigs A/S, Sorø Landevej 302, 4261, Dalmose, Denmark
| | - Nora-Fabienne Paul
- Department of Animal Sciences, Georg-August-University, Burckhardtweg 2, 37077, Göttingen, Germany
| | - Henner Simianer
- Department of Animal Sciences, Georg-August-University, Burckhardtweg 2, 37077, Göttingen, Germany
- Center for Integrated Breeding Research, Georg-August-University, Albrecht-Thaer-Weg 3, 37075, Göttingen, Germany
| | - Jens Tetens
- Department of Animal Sciences, Georg-August-University, Burckhardtweg 2, 37077, Göttingen, Germany
- Center for Integrated Breeding Research, Georg-August-University, Albrecht-Thaer-Weg 3, 37075, Göttingen, Germany
| | - Clemens Falker-Gieske
- Department of Animal Sciences, Georg-August-University, Burckhardtweg 2, 37077, Göttingen, Germany.
- Center for Integrated Breeding Research, Georg-August-University, Albrecht-Thaer-Weg 3, 37075, Göttingen, Germany.
| |
Collapse
|
3
|
Fischer K, Schnieke A. How genome editing changed the world of large animal research. Front Genome Ed 2023; 5:1272687. [PMID: 37886655 PMCID: PMC10598601 DOI: 10.3389/fgeed.2023.1272687] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 09/22/2023] [Indexed: 10/28/2023] Open
Abstract
The first genetically modified large animals were developed in 1985 by microinjection to increase the growth of agricultural livestock such as pigs. Since then, it has been a difficult trail due to the lack of genetic tools. Although methods and technologies were developed quickly for the main experimental mammal, the mouse, e.g., efficient pronuclear microinjection, gene targeting in embryonic stem cells, and omics data, most of it was-and in part still is-lacking when it comes to livestock. Over the next few decades, progress in genetic engineering of large animals was driven less by research for agriculture but more for biomedical applications, such as the production of pharmaceutical proteins in the milk of sheep, goats, or cows, xeno-organ transplantation, and modeling human diseases. Available technologies determined if a desired animal model could be realized, and efficiencies were generally low. Presented here is a short review of how genome editing tools, specifically CRISPR/Cas, have impacted the large animal field in recent years. Although there will be a focus on genome engineering of pigs for biomedical applications, the general principles and experimental approaches also apply to other livestock species or applications.
Collapse
Affiliation(s)
| | - Angelika Schnieke
- Chair of Livestock Biotechnology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| |
Collapse
|
4
|
In vitro genome editing activity of Cas9 in somatic cells after random and transposon-based genomic Cas9 integration. PLoS One 2022; 17:e0279123. [PMID: 36584049 PMCID: PMC9803249 DOI: 10.1371/journal.pone.0279123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 11/29/2022] [Indexed: 12/31/2022] Open
Abstract
Due to its close resemblance, the domesticated pig has proven to be a diverse animal model for biomedical research and genome editing tools have contributed to developing porcine models for several human diseases. By employing the CRISPR-Cas9 system, porcine embryos or somatic cells can be genetically modified to generate the desired genotype. However, somatic cell nuclear transfer (SCNT) of modified somatic cells and embryo manipulation are challenging, especially if the desired genotype is detrimental to the embryo. Direct in vivo edits may facilitate the production of genetically engineered pigs by integrating Cas9 into the porcine genome. Cas9 expressing cells were generated by either random integration or transposon-based integration of Cas9 and used as donor cells in SCNT. In total, 15 animals were generated that carried a transposon-based Cas9 integration and two pigs a randomly integrated Cas9. Cas9 expression was confirmed in muscle, tonsil, spleen, kidney, lymph nodes, oral mucosa, and liver in two boars. Overall, Cas9 expression was higher for transposon-based integration, except in tonsils and liver. To verify Cas9 activity, fibroblasts were subjected to in vitro genome editing. Isolated fibroblasts were transfected with guide RNAs (gRNA) targeting different genes (GGTA1, B4GALNT2, B2M) relevant to xenotransplantation. Next generation sequencing revealed that the editing efficiencies varied (2-60%) between the different target genes. These results show that the integrated Cas9 remained functional, and that Cas9 expressing pigs may be used to induce desired genomic modifications to model human diseases or further evaluate in vivo gene therapy approaches.
Collapse
|
5
|
Eun K, Hwang SU, Kim M, Yoon JD, Kim E, Choi H, Kim G, Jeon HY, Kim JK, Kim JY, Hong N, Park MG, Jang J, Jeong HJ, Kim SJ, Ko BW, Lee SC, Kim H, Hyun SH. Generation of reproductive transgenic pigs of a CRISPR-Cas9-based oncogene-inducible system by somatic cell nuclear transfer. Biotechnol J 2022; 17:e2100434. [PMID: 35233982 DOI: 10.1002/biot.202100434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 01/12/2022] [Accepted: 01/26/2022] [Indexed: 11/06/2022]
Abstract
Alternative cancer models that are close to humans are required to create more valuable preclinical results during oncology studies. Here, we developed a new onco-pig model via developing a CRISPR-Cas9-based Conditional Polycistronic gene expression Cassette (CRI-CPC) system to control the tumor inducing simian virus 40 large T antigen (SV40LT) and oncogenic HRASG12V. After conducting somatic cell nuclear transfer (SCNT), transgenic embryos were transplanted into surrogate mothers and five male piglets were born. Umbilical cord analysis confirmed that all piglets were transgenic. Two of them survived, and they expressed a detectable green fluorescence. We tested whether our CRI-CPC models were naturally fertile and whether the CRI-CPC system was stably transferred to the offspring. By mating with a normal female pig, four offspring piglets were successfully produced. Among them, only three male piglets were transgenic. Finally, we tested their applicability as cancer models after transduction of Cas9 into fibroblasts from each CRI-CPC pig in vitro, resulting in cell acquisition of cancerous characteristics via the induction of oncogene expression. These results showed that our new CRISPR-Cas9-based onco-pig model was successfully developed. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Kiyoung Eun
- Institute of Animal Molecular Biotechnology, Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea.,Department of Biotechnology, School of Life Sciences and Biotechnology, Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Seon-Ung Hwang
- Laboratory of Veterinary Embryology and Biotechnology, Korea University, Seongbuk-gu.,Institute of Stem Cell & Regenerative Medicine, Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju, 28644, Republic of Korea
| | - Mirae Kim
- Laboratory of Veterinary Embryology and Biotechnology, Korea University, Seongbuk-gu.,Institute of Stem Cell & Regenerative Medicine, Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju, 28644, Republic of Korea
| | - Junchul David Yoon
- Laboratory of Veterinary Embryology and Biotechnology, Korea University, Seongbuk-gu.,Institute of Stem Cell & Regenerative Medicine, Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju, 28644, Republic of Korea
| | - Eunhye Kim
- Laboratory of Veterinary Embryology and Biotechnology, Korea University, Seongbuk-gu.,Institute of Stem Cell & Regenerative Medicine, Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju, 28644, Republic of Korea
| | - Hyerin Choi
- Laboratory of Veterinary Embryology and Biotechnology, Korea University, Seongbuk-gu.,Institute of Stem Cell & Regenerative Medicine, Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju, 28644, Republic of Korea
| | - Gahye Kim
- Laboratory of Veterinary Embryology and Biotechnology, Korea University, Seongbuk-gu.,Institute of Stem Cell & Regenerative Medicine, Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju, 28644, Republic of Korea
| | - Hee-Young Jeon
- Institute of Animal Molecular Biotechnology, Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea.,Department of Biotechnology, School of Life Sciences and Biotechnology, Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Jun-Kyum Kim
- Institute of Animal Molecular Biotechnology, Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea.,Department of Biotechnology, School of Life Sciences and Biotechnology, Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Jung Yun Kim
- Institute of Animal Molecular Biotechnology, Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea.,Department of Biotechnology, School of Life Sciences and Biotechnology, Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Nayoung Hong
- Institute of Animal Molecular Biotechnology, Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea.,Department of Biotechnology, School of Life Sciences and Biotechnology, Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Min-Gi Park
- Institute of Animal Molecular Biotechnology, Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea.,Department of Biotechnology, School of Life Sciences and Biotechnology, Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Junseok Jang
- Institute of Animal Molecular Biotechnology, Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea.,Department of Biotechnology, School of Life Sciences and Biotechnology, Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Hyeon Ju Jeong
- Department of Biotechnology, School of Life Sciences and Biotechnology, Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Sung Jin Kim
- Department of Biotechnology, School of Life Sciences and Biotechnology, Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Bong-Woo Ko
- Songbaek Pig Farm, Jeju, 63014, Republic of Korea
| | - Sang Chul Lee
- Cronex Corporation, Cheongju, 28174, Republic of Korea
| | - Hyunggee Kim
- Institute of Animal Molecular Biotechnology, Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea.,Department of Biotechnology, School of Life Sciences and Biotechnology, Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Sang-Hwan Hyun
- Laboratory of Veterinary Embryology and Biotechnology, Korea University, Seongbuk-gu.,Institute of Stem Cell & Regenerative Medicine, Chungbuk National University, 1 Chungdae-ro, Seowon-gu, Cheongju, 28644, Republic of Korea
| |
Collapse
|
6
|
Boettcher AN, Schachtschneider KM, Schook LB, Tuggle CK. Swine models for translational oncological research: an evolving landscape and regulatory considerations. Mamm Genome 2022; 33:230-240. [PMID: 34476572 PMCID: PMC8888764 DOI: 10.1007/s00335-021-09907-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 08/24/2021] [Indexed: 01/19/2023]
Abstract
Swine biomedical models have been gaining in popularity over the last decade, particularly for applications in oncology research. Swine models for cancer research include pigs that have severe combined immunodeficiency for xenotransplantation studies, genetically modified swine models which are capable of developing tumors in vivo, as well as normal immunocompetent pigs. In recent years, there has been a low success rate for the approval of new oncological therapeutics in clinical trials. The two leading reasons for these failures are either due to toxicity and safety issues or lack of efficacy. As all therapeutics must be tested within animal models prior to clinical testing, there are opportunities to expand the ability to assess efficacy and toxicity profiles within the preclinical testing phases of new therapeutics. Most preclinical in vivo testing is performed in mice, canines, and non-human primates. However, swine models are an alternative large animal model for cancer research with similarity to human size, genetics, and physiology. Additionally, tumorigenesis pathways are similar between human and pigs in that similar driver mutations are required for transformation. Due to their larger size, the development of orthotopic tumors is easier than in smaller rodent models; additionally, porcine models can be harnessed for testing of new interventional devices and radiological/surgical approaches as well. Taken together, swine are a feasible option for preclinical therapeutic and device testing. The goals of this resource are to provide a broad overview on regulatory processes required for new therapeutics and devices for use in the clinic, cross-species differences in oncological therapeutic responses, as well as to provide an overview of swine oncology models that have been developed that could be used for preclinical testing to fulfill regulatory requirements.
Collapse
Affiliation(s)
| | - Kyle M. Schachtschneider
- University of Illinois at Chicago, Department of Radiology, Chicago, Illinois, United States,University of Illinois at Urbana-Champaign, National Center for Supercomputing Applications, Urbana, Illinois, United States,University of Illinois at Chicago, Department of Biochemistry and Molecular Genetics, Chicago, Illinois, United States
| | - Lawrence B. Schook
- University of Illinois at Chicago, Department of Radiology, Chicago, Illinois, United States,University of Illinois at Urbana-Champaign, National Center for Supercomputing Applications, Urbana, Illinois, United States,University of Illinois at Urbana-Champaign, Department of Animal Sciences, Illinois, United States
| | - Christopher K Tuggle
- Department of Animal Science, Iowa State University, 2255 Kildee Hall, 806 Stange Road, Ames, IA, 50011, USA.
| |
Collapse
|
7
|
Kalla D, Flisikowski K, Yang K, Sangüesa LB, Kurome M, Kessler B, Zakhartchenko V, Wolf E, Lickert H, Saur D, Schnieke A, Flisikowska T. The Missing Link: Cre Pigs for Cancer Research. Front Oncol 2021; 11:755746. [PMID: 34692545 PMCID: PMC8531543 DOI: 10.3389/fonc.2021.755746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 09/22/2021] [Indexed: 11/13/2022] Open
Abstract
The Cre/loxP system is a powerful tool for the generation of animal models with precise spatial and temporal gene expression. It has proven indispensable in the generation of cancer models with tissue specific expression of oncogenes or the inactivation of tumor suppressor genes. Consequently, Cre-transgenic mice have become an essential prerequisite in basic cancer research. While it is unlikely that pigs will ever replace mice in basic research they are already providing powerful complementary resources for translational studies. But, although conditionally targeted onco-pigs have been generated, no Cre-driver lines exist for any of the major human cancers. To model human pancreatic cancer in pigs, Cre-driver lines were generated by CRISPR/Cas9-mediated insertion of codon-improved Cre (iCre) into the porcine PTF1A gene, thus guaranteeing tissue and cell type specific function which was proven using dual fluorescent reporter pigs. The method used can easily be adapted for the generation of other porcine Cre-driver lines, providing a missing tool for modeling human cancers in large animals.
Collapse
Affiliation(s)
- Daniela Kalla
- Chair of Livestock Biotechnology, Department of Molecular Life Sciences, School of Life Sciences, Technische Universität München, Freising, Germany
| | - Krzysztof Flisikowski
- Chair of Livestock Biotechnology, Department of Molecular Life Sciences, School of Life Sciences, Technische Universität München, Freising, Germany
| | - Kaiyuan Yang
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Munich, Germany
| | - Laura Beltran Sangüesa
- Chair of Livestock Biotechnology, Department of Molecular Life Sciences, School of Life Sciences, Technische Universität München, Freising, Germany
| | - Mayuko Kurome
- Chair of Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Barbara Kessler
- Chair of Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Valeri Zakhartchenko
- Chair of Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Eckhard Wolf
- Chair of Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Munich, Germany
| | - Dieter Saur
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Angelika Schnieke
- Chair of Livestock Biotechnology, Department of Molecular Life Sciences, School of Life Sciences, Technische Universität München, Freising, Germany
| | - Tatiana Flisikowska
- Chair of Livestock Biotechnology, Department of Molecular Life Sciences, School of Life Sciences, Technische Universität München, Freising, Germany
| |
Collapse
|
8
|
Wittayarat M, Hirata M, Namula Z, Sato Y, Nguyen NT, Le QA, Lin Q, Takebayashi K, Tanihara F, Otoi T. Introduction of a point mutation in the KRAS gene of in vitro fertilized porcine zygotes via electroporation of the CRISPR/Cas9 system with single-stranded oligodeoxynucleotides. Anim Sci J 2021; 92:e13534. [PMID: 33638256 DOI: 10.1111/asj.13534] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 11/21/2020] [Accepted: 02/08/2021] [Indexed: 01/02/2023]
Abstract
This study aimed to investigate the efficiency of KRAS gene editing via CRISPR/Cas9 delivery by electroporation and analyzed the effects of the non-homologous end-joining pathway inhibitor Scr7 and single-stranded oligodeoxynucleotide (ssODN) homology arm length on introducing a point mutation in KRAS. Various concentrations (0-2 µM) of Scr7 were evaluated; all concentrations of Scr7 including 0 µM resulted in the generation of blastocysts with a point mutation and the wild-type sequence or indels. No significant differences in the blastocyst formation rates of electroporated zygotes were observed among ssODN homology arm lengths, irrespective of the gRNA (gRNA1 and gRNA2). The proportion of blastocysts carrying a point mutation with or without the wild-type sequence and indels was significantly higher in the ssODN20 group (i.e., the group with a ssODN homology arm of 20 bp) than in the ssODN60 group (gRNA1: 25.7% vs. 5.4% and gRNA2: 45.5% vs. 5.9%, p < .05). In conclusion, the CRISPR/Cas9 delivery with ssODN via electroporation is feasible for the generation of point mutations in porcine embryos. Further studies are required to improve the efficiency and accuracy of the homology-directed repair.
Collapse
Affiliation(s)
- Manita Wittayarat
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan.,Faculty of Veterinary Science, Prince of Songkla University, Songkhla, Thailand
| | - Maki Hirata
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - Zhao Namula
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan.,College of Agricultural Science, Guangdong Ocean University, Guangdong, China
| | - Yoko Sato
- School of Biological Science, Tokai University, Sapporo, Japan
| | - Nhien T Nguyen
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - Quynh A 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
| | - Fuminori Tanihara
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| | - Takeshige Otoi
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima, Japan
| |
Collapse
|
9
|
Yang HJ, Song BS, Sim BW, Jung Y, Chae U, Lee DG, Cha JJ, Baek SJ, Lim KS, Choi WS, Lee HY, Son HC, Park SH, Jeong KJ, Kang P, Baek SH, Koo BS, Kim HN, Jin YB, Park YH, Choo YK, Kim SU. Establishment and Characterization of Immortalized Miniature Pig Pancreatic Cell Lines Expressing Oncogenic K-Ras G12D. Int J Mol Sci 2020; 21:ijms21228820. [PMID: 33233448 PMCID: PMC7700231 DOI: 10.3390/ijms21228820] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/17/2020] [Accepted: 11/19/2020] [Indexed: 12/30/2022] Open
Abstract
In recent decades, many studies on the treatment and prevention of pancreatic cancer have been conducted. However, pancreatic cancer remains incurable, with a high mortality rate. Although mouse models have been widely used for preclinical pancreatic cancer research, these models have many differences from humans. Therefore, large animals may be more useful for the investigation of pancreatic cancer. Pigs have recently emerged as a new model of pancreatic cancer due to their similarities to humans, but no pig pancreatic cancer cell lines have been established for use in drug screening or analysis of tumor biology. Here, we established and characterized an immortalized miniature pig pancreatic cell line derived from primary pancreatic cells and pancreatic cancer-like cells expressing K-rasG12D regulated by the human PTF1A promoter. Using this immortalized cell line, we analyzed the gene expression and phenotypes associated with cancer cell characteristics. Notably, we found that acinar-to-ductal transition was caused by K-rasG12D in the cell line constructed from acinar cells. This may constitute a good research model for the analysis of acinar-to-ductal metaplasia in human pancreatic cancer.
Collapse
Affiliation(s)
- Hae-Jun Yang
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (H.-J.Y.); (B.-S.S.); (B.-W.S.); (Y.J.); (U.C.); (D.G.L.); (J.-J.C.); (S.-J.B.); (K.S.L.); (H.-Y.L.); (H.-C.S.); (P.K.)
- Department of Biological Science, College of Natural Sciences, Wonkwang University, 460, Iksan-daero, Iksan-si 54538, Korea
| | - Bong-Seok Song
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (H.-J.Y.); (B.-S.S.); (B.-W.S.); (Y.J.); (U.C.); (D.G.L.); (J.-J.C.); (S.-J.B.); (K.S.L.); (H.-Y.L.); (H.-C.S.); (P.K.)
| | - Bo-Woong Sim
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (H.-J.Y.); (B.-S.S.); (B.-W.S.); (Y.J.); (U.C.); (D.G.L.); (J.-J.C.); (S.-J.B.); (K.S.L.); (H.-Y.L.); (H.-C.S.); (P.K.)
| | - Yena Jung
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (H.-J.Y.); (B.-S.S.); (B.-W.S.); (Y.J.); (U.C.); (D.G.L.); (J.-J.C.); (S.-J.B.); (K.S.L.); (H.-Y.L.); (H.-C.S.); (P.K.)
| | - Unbin Chae
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (H.-J.Y.); (B.-S.S.); (B.-W.S.); (Y.J.); (U.C.); (D.G.L.); (J.-J.C.); (S.-J.B.); (K.S.L.); (H.-Y.L.); (H.-C.S.); (P.K.)
| | - Dong Gil Lee
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (H.-J.Y.); (B.-S.S.); (B.-W.S.); (Y.J.); (U.C.); (D.G.L.); (J.-J.C.); (S.-J.B.); (K.S.L.); (H.-Y.L.); (H.-C.S.); (P.K.)
| | - Jae-Jin Cha
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (H.-J.Y.); (B.-S.S.); (B.-W.S.); (Y.J.); (U.C.); (D.G.L.); (J.-J.C.); (S.-J.B.); (K.S.L.); (H.-Y.L.); (H.-C.S.); (P.K.)
| | - Seo-Jong Baek
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (H.-J.Y.); (B.-S.S.); (B.-W.S.); (Y.J.); (U.C.); (D.G.L.); (J.-J.C.); (S.-J.B.); (K.S.L.); (H.-Y.L.); (H.-C.S.); (P.K.)
| | - Kyung Seob Lim
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (H.-J.Y.); (B.-S.S.); (B.-W.S.); (Y.J.); (U.C.); (D.G.L.); (J.-J.C.); (S.-J.B.); (K.S.L.); (H.-Y.L.); (H.-C.S.); (P.K.)
| | - Won Seok Choi
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (W.S.C.); (S.-H.P.); (K.-J.J.); (S.H.B.); (B.-S.K.); (H.-N.K.); (Y.B.J.)
| | - Hwal-Yong Lee
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (H.-J.Y.); (B.-S.S.); (B.-W.S.); (Y.J.); (U.C.); (D.G.L.); (J.-J.C.); (S.-J.B.); (K.S.L.); (H.-Y.L.); (H.-C.S.); (P.K.)
| | - Hee-Chang Son
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (H.-J.Y.); (B.-S.S.); (B.-W.S.); (Y.J.); (U.C.); (D.G.L.); (J.-J.C.); (S.-J.B.); (K.S.L.); (H.-Y.L.); (H.-C.S.); (P.K.)
| | - Sung-Hyun Park
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (W.S.C.); (S.-H.P.); (K.-J.J.); (S.H.B.); (B.-S.K.); (H.-N.K.); (Y.B.J.)
| | - Kang-Jin Jeong
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (W.S.C.); (S.-H.P.); (K.-J.J.); (S.H.B.); (B.-S.K.); (H.-N.K.); (Y.B.J.)
| | - Philyong Kang
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (H.-J.Y.); (B.-S.S.); (B.-W.S.); (Y.J.); (U.C.); (D.G.L.); (J.-J.C.); (S.-J.B.); (K.S.L.); (H.-Y.L.); (H.-C.S.); (P.K.)
| | - Seung Ho Baek
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (W.S.C.); (S.-H.P.); (K.-J.J.); (S.H.B.); (B.-S.K.); (H.-N.K.); (Y.B.J.)
| | - Bon-Sang Koo
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (W.S.C.); (S.-H.P.); (K.-J.J.); (S.H.B.); (B.-S.K.); (H.-N.K.); (Y.B.J.)
| | - Han-Na Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (W.S.C.); (S.-H.P.); (K.-J.J.); (S.H.B.); (B.-S.K.); (H.-N.K.); (Y.B.J.)
| | - Yeung Bae Jin
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (W.S.C.); (S.-H.P.); (K.-J.J.); (S.H.B.); (B.-S.K.); (H.-N.K.); (Y.B.J.)
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, 501 Jinjudaero, Jinju 52828, Korea
| | - Young-Ho Park
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (H.-J.Y.); (B.-S.S.); (B.-W.S.); (Y.J.); (U.C.); (D.G.L.); (J.-J.C.); (S.-J.B.); (K.S.L.); (H.-Y.L.); (H.-C.S.); (P.K.)
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34113, Korea
- Correspondence: (Y.-H.P.); (Y.-K.C.); (S.-U.K.); Tel.: +82-43-240-6321 (S.-U.K.); Fax: +82-43-240-6309 (S.-U.K.)
| | - Young-Kug Choo
- Department of Biological Science, College of Natural Sciences, Wonkwang University, 460, Iksan-daero, Iksan-si 54538, Korea
- Correspondence: (Y.-H.P.); (Y.-K.C.); (S.-U.K.); Tel.: +82-43-240-6321 (S.-U.K.); Fax: +82-43-240-6309 (S.-U.K.)
| | - Sun-Uk Kim
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju-si 28116, Korea; (H.-J.Y.); (B.-S.S.); (B.-W.S.); (Y.J.); (U.C.); (D.G.L.); (J.-J.C.); (S.-J.B.); (K.S.L.); (H.-Y.L.); (H.-C.S.); (P.K.)
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34113, Korea
- Correspondence: (Y.-H.P.); (Y.-K.C.); (S.-U.K.); Tel.: +82-43-240-6321 (S.-U.K.); Fax: +82-43-240-6309 (S.-U.K.)
| |
Collapse
|
10
|
Saricaoglu ÖC, Teller S, Wang X, Wang S, Stupakov P, Heinrich T, Istvanffy R, Friess H, Ceyhan GO, Demir IE. Localisation analysis of nerves in the mouse pancreas reveals the sites of highest nerve density and nociceptive innervation. Neurogastroenterol Motil 2020; 32:e13880. [PMID: 32406093 DOI: 10.1111/nmo.13880] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 03/03/2020] [Accepted: 04/17/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Neuropathy and neuro-inflammation drive the severe pain and disease progression in human chronic pancreatitis and pancreatic cancer. Mice, especially genetically induced-mouse models, have been increasingly utilized in mechanistic research on pancreatic neuropathy, but the normal "peripheral neurobiology" of the mouse pancreas has not yet been critically compared to human pancreas. METHODS We introduced a standardized tissue-harvesting technique that preserves the anatomic orientation of the mouse pancreas and allows complete sectioning in an anterior to posterior fashion. We applied immunohistochemistry and quantitative colorimetry of all nerves from the whole organ for studying pancreatic neuro-anatomy. KEY RESULTS Nerves in the mouse pancreas appeared as "clusters" of nerve trunks in contrast to singly distributed nerve trunks in the human pancreas. Nerve trunks in the mouse pancreas were exclusively found around intrapancreatic blood vessels, and around lymphoid structures. The majority of nerve trunks were located in the pancreatic head (0.15 ± 0.08% of tissue area) and the anterior/front surface of the corpus/body (0.17 ± 0.27%), thus significantly more than in the tail (0.02 ± 0.02%, P = .006). Nerves in the tail included a higher proportion of nociceptive fibers, but the absolute majority, ie, ca. 70%, of all nociceptive fibers, were localized in the head. Mice heterozygous for Bdnf knockout allele (Bdnf+/- ) exhibited enrichment of nitrergic nerve fibers specifically in the head and corpus. CONCLUSIONS & INFERENCES Neuro-anatomy of the "mesenteric type" mouse pancreas is highly different from the "compact" human pancreas. Studies that aim at reproducing human pancreatic neuro-phenomena in mouse models should pay diligent attention to these anatomic differences.
Collapse
Affiliation(s)
- Ömer Cemil Saricaoglu
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Steffen Teller
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Xiaobo Wang
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Shenghan Wang
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Pavel Stupakov
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Tobias Heinrich
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Rouzanna Istvanffy
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Helmut Friess
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Güralp O Ceyhan
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany.,Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Ihsan Ekin Demir
- Department of Surgery, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany.,Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey.,German Cancer Consortium (DKTK), Partner Site, Munich, Germany.,CRC 1321 Modelling and Targeting Pancreatic Cancer, Munich, Germany
| |
Collapse
|
11
|
Kalla D, Kind A, Schnieke A. Genetically Engineered Pigs to Study Cancer. Int J Mol Sci 2020; 21:E488. [PMID: 31940967 PMCID: PMC7013672 DOI: 10.3390/ijms21020488] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 02/06/2023] Open
Abstract
Recent decades have seen groundbreaking advances in cancer research. Genetically engineered animal models, mainly in mice, have contributed to a better understanding of the underlying mechanisms involved in cancer. However, mice are not ideal for translating basic research into studies closer to the clinic. There is a need for complementary information provided by non-rodent species. Pigs are well suited for translational biomedical research as they share many similarities with humans such as body and organ size, aspects of anatomy, physiology and pathophysiology and can provide valuable means of developing and testing novel diagnostic and therapeutic procedures. Porcine oncology is a new field, but it is clear that replication of key oncogenic mutation in pigs can usefully mimic several human cancers. This review briefly outlines the technology used to generate genetically modified pigs, provides an overview of existing cancer models, their applications and how the field may develop in the near future.
Collapse
Affiliation(s)
| | | | - Angelika Schnieke
- Chair of Livestock Biotechnology, School of Life Sciences, Technische Universität München, 85354 Freising, Germany; (D.K.); (A.K.)
| |
Collapse
|
12
|
Overgaard NH, Fan TM, Schachtschneider KM, Principe DR, Schook LB, Jungersen G. Of Mice, Dogs, Pigs, and Men: Choosing the Appropriate Model for Immuno-Oncology Research. ILAR J 2019; 59:247-262. [PMID: 30476148 DOI: 10.1093/ilar/ily014] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 07/30/2018] [Indexed: 02/06/2023] Open
Abstract
The immune system plays dual roles in response to cancer. The host immune system protects against tumor formation via immunosurveillance; however, recognition of the tumor by immune cells also induces sculpting mechanisms leading to a Darwinian selection of tumor cell variants with reduced immunogenicity. Cancer immunoediting is the concept used to describe the complex interplay between tumor cells and the immune system. This concept, commonly referred to as the three E's, is encompassed by 3 distinct phases of elimination, equilibrium, and escape. Despite impressive results in the clinic, cancer immunotherapy still has room for improvement as many patients remain unresponsive to therapy. Moreover, many of the preclinical results obtained in the widely used mouse models of cancer are lost in translation to human patients. To improve the success rate of immuno-oncology research and preclinical testing of immune-based anticancer therapies, using alternative animal models more closely related to humans is a promising approach. Here, we describe 2 of the major alternative model systems: canine (spontaneous) and porcine (experimental) cancer models. Although dogs display a high rate of spontaneous tumor formation, an increased number of genetically modified porcine models exist. We suggest that the optimal immuno-oncology model may depend on the stage of cancer immunoediting in question. In particular, the spontaneous canine tumor models provide a unique platform for evaluating therapies aimed at the escape phase of cancer, while genetically engineered swine allow for elucidation of tumor-immune cell interactions especially during the phases of elimination and equilibrium.
Collapse
Affiliation(s)
- Nana H Overgaard
- Department of Micro- and Nanotechnology, Technical University of Denmark, Kgs Lyngby, Denmark
| | - Timothy M Fan
- Department of Veterinary Clinical Medicine, University of Illinois, Urbana-Champaign, Illinois
| | | | - Daniel R Principe
- Medical Scientist Training Program, University of Illinois College of Medicine, Chicago, Illinois
| | - Lawrence B Schook
- Department of Radiology, University of Illinois, Chicago, Illinois.,Department of Animal Sciences, University of Illinois, Urbana-Champaign, Illinois
| | - Gregers Jungersen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| |
Collapse
|
13
|
Rossa C, D'Silva NJ. Non-murine models to investigate tumor-immune interactions in head and neck cancer. Oncogene 2019; 38:4902-4914. [PMID: 30872793 PMCID: PMC6586515 DOI: 10.1038/s41388-019-0776-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/24/2019] [Accepted: 02/25/2019] [Indexed: 12/18/2022]
Abstract
The immune response has important roles in the biology of solid tumors, including oncogenesis, tumor growth, invasion and metastasis, and response to treatment. Improved understanding of tumor-immune system interactions has provided promising therapeutic options that are based on the rescue and enhancement of the anti-tumoral host response. Immune-based treatments have been approved for clinical use in various types of cancer, including head and neck cancer (HNC); other strategies involving combination therapies are currently in development. These novel therapies were developed based on knowledge derived from in vitro, in silico, and in vivo pre-clinical studies. However, clinical trials seldom replicate the efficacy observed in pre-clinical animal studies. This lack of correlation between pre-clinical studies and clinical trials may be related to limitations of the models used; which highlights the relevance of considering immune-related aspects of different pre-clinical models. Murine models are the most frequently used pre-clinical models of HNC and are discussed elsewhere. Non-murine models have characteristics that offer unique opportunities for the study of HNC etiology, therapeutic strategies, and tumor-immune system interactions. The current review focuses on immune-related aspects of non-murine models, including dog, cat, pig, zebrafish, and frog, that could be used to investigate tumor-immune interactions in HNC.
Collapse
Affiliation(s)
- Carlos Rossa
- Department of Diagnosis and Surgery, UNESP-State University of Sao Paulo, School of Dentistry at Araraquara, Araraquara, SP, Brazil. .,Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA.
| | - Nisha J D'Silva
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA. .,Department of Pathology, Medical School, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
14
|
Boettcher AN, Kiupel M, Adur MK, Cocco E, Santin AD, Bellone S, Charley SE, Blanco-Fernandez B, Risinger JI, Ross JW, Tuggle CK, Shapiro EM. Human Ovarian Cancer Tumor Formation in Severe Combined Immunodeficient (SCID) Pigs. Front Oncol 2019; 9:9. [PMID: 30723704 PMCID: PMC6349777 DOI: 10.3389/fonc.2019.00009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Accepted: 01/03/2019] [Indexed: 01/07/2023] Open
Abstract
Ovarian cancer (OvCa) is the most lethal gynecologic malignancy, with two-thirds of patients having late-stage disease (II-IV) at diagnosis. Improved diagnosis and therapies are needed, yet preclinical animal models for ovarian cancer research have primarily been restricted to rodents, for data on which can fail to translate to the clinic. Thus, there is currently a need for a large animal OvCa model. Therefore, we sought to determine if pigs, being more similar to humans in terms of anatomy and physiology, would be a viable preclinical animal model for OvCa. We injected human OSPC-ARK1 cells, a chemotherapy-resistant primary ovarian serous papillary carcinoma cell line, into the neck muscle and ear tissue of four severe combined immune deficient (SCID) and two non-SCID pigs housed in novel biocontainment facilities to study the ability of human OvCa cells to form tumors in a xenotransplantation model. Tumors developed in ear tissue of three SCID pigs, while two SCID pigs developed tumors in neck tissue; no tumors were detected in non-SCID control pigs. All tumor masses were confirmed microscopically as ovarian carcinomas. The carcinomas in SCID pigs were morphologically similar to the original ovarian carcinoma and had the same immunohistochemical phenotype based on expression of Claudin 3, Claudin 4, Cytokeratin 7, p16, and EMA. Confirmation that OSPC-ARK1 cells form carcinomas in SCID pigs substantiates further development of orthotopic models of OvCa in pigs.
Collapse
Affiliation(s)
- Adeline N Boettcher
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Matti Kiupel
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI, United States
| | - Malavika K Adur
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Emiliano Cocco
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, United States.,Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Alessandro D Santin
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, United States
| | - Stefania Bellone
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, United States
| | - Sara E Charley
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | | | - John I Risinger
- Department of Radiology, Michigan State University, East Lansing, MI, United States.,Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI, United States
| | - Jason W Ross
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | | | - Erik M Shapiro
- Department of Radiology, Michigan State University, East Lansing, MI, United States.,Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
15
|
Boettcher AN, Loving CL, Cunnick JE, Tuggle CK. Development of Severe Combined Immunodeficient (SCID) Pig Models for Translational Cancer Modeling: Future Insights on How Humanized SCID Pigs Can Improve Preclinical Cancer Research. Front Oncol 2018; 8:559. [PMID: 30560086 PMCID: PMC6284365 DOI: 10.3389/fonc.2018.00559] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 11/09/2018] [Indexed: 12/13/2022] Open
Abstract
Within the last decade there have been several severe combined immunodeficient (SCID) pig models discovered or genetically engineered. The animals have mutations in ARTEMIS, IL2RG, or RAG1/2 genes, or combinations thereof, providing SCID pigs with NK cells, but deficient in T and B cells, or deficient in NK, T, and B cells for research studies. Biocontainment facilities and positive pressure isolators are developed to limit pathogen exposure and prolong the life of SCID pigs. Raising SCID pigs in such facilities allows for completion of long-term studies such as xenotransplantation of human cells. Ectopically injected human cancer cell lines develop into tumors in SCID pigs, thus providing a human-sized in vivo model for evaluating imaging methods to improve cancer detection and therapeutic research and development. Immunocompromised pigs have the potential to be immunologically humanized by xenotransplantation with human hematopoietic stem cells, peripheral blood leukocytes, or fetal tissue. These cells can be introduced through various routes including injection into fetal liver or the intraperitoneal (IP) space, or into piglets by intravenous, IP, and intraosseous administration. The development and maintenance of transplanted human immune cells would be initially (at least) dependent on immune signaling from swine cells. Compared to mice, swine share higher homology in immune related genes with humans. We hypothesize that the SCID pig may be able to support improved engraftment and differentiation of a wide range of human immune cells as compared to equivalent mouse models. Humanization of SCID pigs would thus provide a valuable model system for researchers to study interactions between human tumor and human immune cells. Additionally, as the SCID pig model is further developed, it may be possible to develop patient-derived xenograft models for individualized therapy and drug testing. We thus theorize that the individualized therapeutic approach would be significantly improved with a humanized SCID pig due to similarities in size, metabolism, and physiology. In all, porcine SCID models have significant potential as an excellent preclinical animal model for therapeutic testing.
Collapse
Affiliation(s)
| | - Crystal L. Loving
- Food Safety and Enteric Pathogens Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, United States
| | - Joan E. Cunnick
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | | |
Collapse
|
16
|
Perleberg C, Kind A, Schnieke A. Genetically engineered pigs as models for human disease. Dis Model Mech 2018; 11:11/1/dmm030783. [PMID: 29419487 PMCID: PMC5818075 DOI: 10.1242/dmm.030783] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Genetically modified animals are vital for gaining a proper understanding of disease mechanisms. Mice have long been the mainstay of basic research into a wide variety of diseases but are not always the most suitable means of translating basic knowledge into clinical application. The shortcomings of rodent preclinical studies are widely recognised, and regulatory agencies around the world now require preclinical trial data from nonrodent species. Pigs are well suited to biomedical research, sharing many similarities with humans, including body size, anatomical features, physiology and pathophysiology, and they already play an important role in translational studies. This role is set to increase as advanced genetic techniques simplify the generation of pigs with precisely tailored modifications designed to replicate lesions responsible for human disease. This article provides an overview of the most promising and clinically relevant genetically modified porcine models of human disease for translational biomedical research, including cardiovascular diseases, cancers, diabetes mellitus, Alzheimer's disease, cystic fibrosis and Duchenne muscular dystrophy. We briefly summarise the technologies involved and consider the future impact of recent technical advances. Summary: An overview of porcine models of human disease, including cardiovascular diseases, cancers, diabetes mellitus, Alzheimer's disease, cystic fibrosis and Duchenne muscular dystrophy. We summarise the technologies involved and potential future impact of recent technical advances.
Collapse
Affiliation(s)
- Carolin Perleberg
- Chair of Livestock Biotechnology, School of Life Sciences, Technische Universität München, 85354 Freising, Germany
| | - Alexander Kind
- Chair of Livestock Biotechnology, School of Life Sciences, Technische Universität München, 85354 Freising, Germany
| | - Angelika Schnieke
- Chair of Livestock Biotechnology, School of Life Sciences, Technische Universität München, 85354 Freising, Germany
| |
Collapse
|
17
|
Marx H, Hahne H, Ulbrich SE, Schnieke A, Rottmann O, Frishman D, Kuster B. Annotation of the Domestic Pig Genome by Quantitative Proteogenomics. J Proteome Res 2017. [PMID: 28625053 DOI: 10.1021/acs.jproteome.7b00184] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The pig is one of the earliest domesticated animals in the history of human civilization and represents one of the most important livestock animals. The recent sequencing of the Sus scrofa genome was a major step toward the comprehensive understanding of porcine biology, evolution, and its utility as a promising large animal model for biomedical and xenotransplantation research. However, the functional and structural annotation of the Sus scrofa genome is far from complete. Here, we present mass spectrometry-based quantitative proteomics data of nine juvenile organs and six embryonic stages between 18 and 39 days after gestation. We found that the data provide evidence for and improve the annotation of 8176 protein-coding genes including 588 novel and 321 refined gene models. The analysis of tissue-specific proteins and the temporal expression profiles of embryonic proteins provides an initial functional characterization of expressed protein interaction networks and modules including as yet uncharacterized proteins. Comparative transcript and protein expression analysis to human organs reveal a moderate conservation of protein translation across species. We anticipate that this resource will facilitate basic and applied research on Sus scrofa as well as its porcine relatives.
Collapse
Affiliation(s)
| | | | | | | | | | - Dmitrij Frishman
- Institute of Bioinformatics and Systems Biology , German Research Center for Environmental Health, Neuherberg, Germany.,St Petersburg State Polytechnical University , St Petersburg, Russia
| | - Bernhard Kuster
- Center for Integrated Protein Science Munich , Munich, Germany
| |
Collapse
|
18
|
Flisikowska T, Kind A, Schnieke A. Pigs as models of human cancers. Theriogenology 2016; 86:433-7. [DOI: 10.1016/j.theriogenology.2016.04.058] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 03/14/2016] [Indexed: 10/21/2022]
|
19
|
Watson AL, Carlson DF, Largaespada DA, Hackett PB, Fahrenkrug SC. Engineered Swine Models of Cancer. Front Genet 2016; 7:78. [PMID: 27242889 PMCID: PMC4860525 DOI: 10.3389/fgene.2016.00078] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 04/18/2016] [Indexed: 12/13/2022] Open
Abstract
Over the past decade, the technology to engineer genetically modified swine has seen many advancements, and because their physiology is remarkably similar to that of humans, swine models of cancer may be extremely valuable for preclinical safety studies as well as toxicity testing of pharmaceuticals prior to the start of human clinical trials. Hence, the benefits of using swine as a large animal model in cancer research and the potential applications and future opportunities of utilizing pigs in cancer modeling are immense. In this review, we discuss how pigs have been and can be used as a biomedical models for cancer research, with an emphasis on current technologies. We have focused on applications of precision genetics that can provide models that mimic human cancer predisposition syndromes. In particular, we describe the advantages of targeted gene-editing using custom endonucleases, specifically TALENs and CRISPRs, and transposon systems, to make novel pig models of cancer with broad preclinical applications.
Collapse
Affiliation(s)
| | | | - David A Largaespada
- RecombineticsSt. Paul, MN, USA; Masonic Cancer Center, University of MinnesotaMinneapolis, MN, USA; Genetics, Cell Biology and Development, University of MinnesotaMinneapolis, MN, USA; Pediatrics, University of MinnesotaMinneapolis, MN, USA
| | - Perry B Hackett
- RecombineticsSt. Paul, MN, USA; Genetics, Cell Biology and Development, University of MinnesotaMinneapolis, MN, USA; Center for Genome Engineering, University of MinnesotaMinneapolis, MN, USA
| | | |
Collapse
|
20
|
Saalfrank A, Janssen KP, Ravon M, Flisikowski K, Eser S, Steiger K, Flisikowska T, Müller-Fliedner P, Schulze É, Brönner C, Gnann A, Kappe E, Böhm B, Schade B, Certa U, Saur D, Esposito I, Kind A, Schnieke A. A porcine model of osteosarcoma. Oncogenesis 2016; 5:e210. [PMID: 26974205 PMCID: PMC4815050 DOI: 10.1038/oncsis.2016.19] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 01/25/2016] [Accepted: 01/28/2016] [Indexed: 12/15/2022] Open
Abstract
We previously produced pigs with a latent oncogenic TP53 mutation. Humans with TP53 germline mutations are predisposed to a wide spectrum of early-onset cancers, predominantly breast, brain, adrenal gland cancer, soft tissue sarcomas and osteosarcomas. Loss of p53 function has been observed in >50% of human cancers. Here we demonstrate that porcine mesenchymal stem cells (MSCs) convert to a transformed phenotype after activation of latent oncogenic TP53R167H and KRASG12D, and overexpression of MYC promotes tumorigenesis. The process mimics key molecular aspects of human sarcomagenesis. Transformed porcine MSCs exhibit genomic instability, with complex karyotypes, and develop into sarcomas on transplantation into immune-deficient mice. In pigs, heterozygous knockout of TP53 was sufficient for spontaneous osteosarcoma development in older animals, whereas homozygous TP53 knockout resulted in multiple large osteosarcomas in 7–8-month-old animals. This is the first report that engineered mutation of an endogenous tumour-suppressor gene leads to invasive cancer in pigs. Unlike in Trp53 mutant mice, osteosarcoma developed in the long bones and skull, closely recapitulating the human disease. These animals thus promise a model for juvenile osteosarcoma, a relatively uncommon but devastating disease.
Collapse
Affiliation(s)
- A Saalfrank
- Chair of Livestock Biotechnology, Technische Universität München, Freising, Germany
| | - K-P Janssen
- Department of Surgery, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - M Ravon
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - K Flisikowski
- Chair of Livestock Biotechnology, Technische Universität München, Freising, Germany
| | - S Eser
- Department of Medicine II, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - K Steiger
- Department of Pathology, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - T Flisikowska
- Chair of Livestock Biotechnology, Technische Universität München, Freising, Germany
| | - P Müller-Fliedner
- Chair of Livestock Biotechnology, Technische Universität München, Freising, Germany
| | - É Schulze
- Chair of Livestock Biotechnology, Technische Universität München, Freising, Germany
| | - C Brönner
- Chair of Livestock Biotechnology, Technische Universität München, Freising, Germany
| | - A Gnann
- Department of Surgery, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - E Kappe
- Department of Pathology, Bavarian Animal Health Service, Poing, Germany
| | - B Böhm
- Department of Pathology, Bavarian Animal Health Service, Poing, Germany
| | - B Schade
- Department of Pathology, Bavarian Animal Health Service, Poing, Germany
| | - U Certa
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - D Saur
- Department of Medicine II, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - I Esposito
- Institute of Pathology, Heinrich-Heine-University of Düsseldorf, Düsseldorf, Germany
| | - A Kind
- Chair of Livestock Biotechnology, Technische Universität München, Freising, Germany
| | - A Schnieke
- Chair of Livestock Biotechnology, Technische Universität München, Freising, Germany
| |
Collapse
|
21
|
Schook LB, Rund L, Begnini KR, Remião MH, Seixas FK, Collares T. Emerging Technologies to Create Inducible and Genetically Defined Porcine Cancer Models. Front Genet 2016; 7:28. [PMID: 26973698 PMCID: PMC4770043 DOI: 10.3389/fgene.2016.00028] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 02/13/2016] [Indexed: 12/26/2022] Open
Abstract
There is an emerging need for new animal models that address unmet translational cancer research requirements. Transgenic porcine models provide an exceptional opportunity due to their genetic, anatomic, and physiological similarities with humans. Due to recent advances in the sequencing of domestic animal genomes and the development of new organism cloning technologies, it is now very feasible to utilize pigs as a malleable species, with similar anatomic and physiological features with humans, in which to develop cancer models. In this review, we discuss genetic modification technologies successfully used to produce porcine biomedical models, in particular the Cre-loxP System as well as major advances and perspectives the CRISPR/Cas9 System. Recent advancements in porcine tumor modeling and genome editing will bring porcine models to the forefront of translational cancer research.
Collapse
Affiliation(s)
- Lawrence B Schook
- University of Illinois Cancer Center, University of Illinois at ChicagoChicago, IL, USA; Department of Animal Sciences, University of Illinois at Urbana-ChampaignChampaign, IL, USA
| | - Laurie Rund
- Department of Animal Sciences, University of Illinois at Urbana-Champaign Champaign, IL, USA
| | - Karine R Begnini
- Postgraduate Program in Biotechnology, Biotechnology Unit, Technology Development Center, Federal University of Pelotas Pelotas, Brazil
| | - Mariana H Remião
- Postgraduate Program in Biotechnology, Biotechnology Unit, Technology Development Center, Federal University of Pelotas Pelotas, Brazil
| | - Fabiana K Seixas
- Postgraduate Program in Biotechnology, Biotechnology Unit, Technology Development Center, Federal University of Pelotas Pelotas, Brazil
| | - Tiago Collares
- Postgraduate Program in Biotechnology, Biotechnology Unit, Technology Development Center, Federal University of Pelotas Pelotas, Brazil
| |
Collapse
|
22
|
Genetically engineered livestock for biomedical models. Transgenic Res 2016; 25:345-59. [PMID: 26820410 DOI: 10.1007/s11248-016-9928-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 01/06/2016] [Indexed: 12/23/2022]
Abstract
To commemorate Transgenic Animal Research Conference X, this review summarizes the recent progress in developing genetically engineered livestock species as biomedical models. The first of these conferences was held in 1997, which turned out to be a watershed year for the field, with two significant events occurring. One was the publication of the first transgenic livestock animal disease model, a pig with retinitis pigmentosa. Before that, the use of livestock species in biomedical research had been limited to wild-type animals or disease models that had been induced or were naturally occurring. The second event was the report of Dolly, a cloned sheep produced by somatic cell nuclear transfer. Cloning subsequently became an essential part of the process for most of the models developed in the last 18 years and is stilled used prominently today. This review is intended to highlight the biomedical modeling achievements that followed those key events, many of which were first reported at one of the previous nine Transgenic Animal Research Conferences. Also discussed are the practical challenges of utilizing livestock disease models now that the technical hurdles of model development have been largely overcome.
Collapse
|
23
|
Abstract
The large size of the pig and its similarity in anatomy, physiology, metabolism, and genetics to humans make it an ideal platform to develop a genetically defined, large animal model of cancer. To this end, we created a transgenic “oncopig” line encoding Cre recombinase inducible porcine transgenes encoding KRASG12D and TP53R167H, which represent a commonly mutated oncogene and tumor suppressor in human cancers, respectively. Treatment of cells derived from these oncopigs with the adenovirus encoding Cre (AdCre) led to KRASG12D and TP53R167H expression, which rendered the cells transformed in culture and tumorigenic when engrafted into immunocompromised mice. Finally, injection of AdCre directly into these oncopigs led to the rapid and reproducible tumor development of mesenchymal origin. Transgenic animals receiving AdGFP (green fluorescent protein) did not have any tumor mass formation or altered histopathology. This oncopig line could thus serve as a genetically malleable model for potentially a wide spectrum of cancers, while controlling for temporal or spatial genesis, which should prove invaluable to studies previously hampered by the lack of a large animal model of cancer.
Collapse
|