1
|
Blanchard Z, Brown EA, Ghazaryan A, Welm AL. PDX models for functional precision oncology and discovery science. Nat Rev Cancer 2024:10.1038/s41568-024-00779-3. [PMID: 39681638 DOI: 10.1038/s41568-024-00779-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/19/2024] [Indexed: 12/18/2024]
Abstract
Precision oncology relies on detailed molecular analysis of how diverse tumours respond to various therapies, with the aim to optimize treatment outcomes for individual patients. Patient-derived xenograft (PDX) models have been key to preclinical validation of precision oncology approaches, enabling the analysis of each tumour's unique genomic landscape and testing therapies that are predicted to be effective based on specific mutations, gene expression patterns or signalling abnormalities. To extend these standard precision oncology approaches, the field has strived to complement the otherwise static and often descriptive measurements with functional assays, termed functional precision oncology (FPO). By utilizing diverse PDX and PDX-derived models, FPO has gained traction as an effective preclinical and clinical tool to more precisely recapitulate patient biology using in vivo and ex vivo functional assays. Here, we explore advances and limitations of PDX and PDX-derived models for precision oncology and FPO. We also examine the future of PDX models for precision oncology in the age of artificial intelligence. Integrating these two disciplines could be the key to fast, accurate and cost-effective treatment prediction, revolutionizing oncology and providing patients with cancer with the most effective, personalized treatments.
Collapse
Affiliation(s)
- Zannel Blanchard
- Department of Oncological Sciences, University of Utah, Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Elisabeth A Brown
- Department of Oncological Sciences, University of Utah, Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Arevik Ghazaryan
- Department of Oncological Sciences, University of Utah, Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Alana L Welm
- Department of Oncological Sciences, University of Utah, Huntsman Cancer Institute, Salt Lake City, UT, USA.
| |
Collapse
|
2
|
Monarch K, Yoon J, Uh K, Reese E, Restrepo DC, de Carvalho Madrid DM, Touchard L, Spate LD, Samuel MS, Driver JP, Lim JH, Schlink S, Whitworth KM, Wells KD, Prather RS, Chen PR, Lee K. Fetal bone engraftment reconstitutes the immune system in pigs with severe combined immunodeficiency. Lab Anim (NY) 2024; 53:276-286. [PMID: 39289566 PMCID: PMC11439731 DOI: 10.1038/s41684-024-01439-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/21/2024] [Indexed: 09/19/2024]
Abstract
Genetic modification of genes such as recombination activating gene 2 (RAG2) or interleukin-2 receptor-γ (IL2RG) results in pigs exhibiting severe combined immunodeficiency (SCID). Pigs presenting a SCID phenotype are important animal models that can be used to establish xenografts and to study immune system development and various immune-related pathologies. However, due to their immunocompromised nature, SCID pigs have shortened lifespans and are notoriously difficult to maintain. The failure-to-thrive phenotype makes the establishment of a breeding population of RAG2/IL2RG double-knockout pigs virtually impossible. Here, to overcome this limitation, we investigated whether reconstituting the immune system of SCID piglets with a fetal bone allograft would extend their lifespan. Following intramuscular transplantation, allografts gave rise to lymphocytes expressing T cell (CD3, CD4 and CD8), B cell (CD79α) and natural killer cell (CD335) lineage markers, which were detected in circulation as well in the spleen, liver, bone marrow and thymic tissues. The presence of lymphocytes indicates broad engraftment of donor cells in the recipient SCID pigs. Unlike unreconstituted SCID pigs, the engrafted animals thrived and reached puberty under standard housing conditions. This study demonstrates a novel method to extend the survival of SCID pigs, which may improve the availability and use of SCID pigs as a biomedical animal model.
Collapse
Affiliation(s)
- Kaylynn Monarch
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, Columbia, MO, USA
| | - Junchul Yoon
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, Columbia, MO, USA
| | - Kyungjun Uh
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Emily Reese
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, Columbia, MO, USA
| | - Diana Canaveral Restrepo
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, Columbia, MO, USA
| | | | - Laurie Touchard
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, Columbia, MO, USA
| | - Lee D Spate
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, Columbia, MO, USA
| | - Melissa S Samuel
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, Columbia, MO, USA
- National Swine Resource and Research Center, University of Missouri, Columbia, MO, USA
| | - John P Driver
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, Columbia, MO, USA
| | - Ji-Hey Lim
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Sarah Schlink
- Office of Animal Resources, University of Missouri, Columbia, MO, USA
| | - Kristin M Whitworth
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, Columbia, MO, USA
- National Swine Resource and Research Center, University of Missouri, Columbia, MO, USA
| | - Kevin D Wells
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, Columbia, MO, USA
- National Swine Resource and Research Center, University of Missouri, Columbia, MO, USA
| | - Randall S Prather
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, Columbia, MO, USA
- National Swine Resource and Research Center, University of Missouri, Columbia, MO, USA
| | - Paula R Chen
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, Columbia, MO, USA.
- United States Department of Agriculture - Agriculture Research Service, Plant Genetics Research Unit, Columbia, MO, USA.
| | - Kiho Lee
- Division of Animal Science, College of Agriculture Food and Natural Resources, University of Missouri, Columbia, MO, USA.
- National Swine Resource and Research Center, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
3
|
Wang J, Zhu F, Jiao D, Yang C, Wang J, Wang F, Zhao H, Wei HJ, Zhao HY. Generation of RAG2 Knockout Immune-Deficient Miniature Pigs. Animals (Basel) 2024; 14:2597. [PMID: 39272382 PMCID: PMC11393836 DOI: 10.3390/ani14172597] [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: 08/05/2024] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
Recombination-activating genes (RAGs) play a crucial role in the V(D)J recombination process and the development of immune cells. The development of the immune system and its mechanisms in pigs exhibit greater similarity to those of humans compared to other animals, thus rendering pigs a valuable tool for biomedical research. In this study, we utilized CRISPR/Cas9 gene editing and somatic cell nuclear transfer technology to generate RAG2 knockout (KO) pigs. Furthermore, we evaluated the impact of RAG2 KO on the immune organs and immune cell development through morphological observations, blood analysis and flow cytometry technology. RAG2 KO cell lines were used as donors for cloning. The reconstructed embryos were transplanted into 4 surrogate sows, and after 116 days of gestation, 2 sows gave birth to 12 live piglets, all of which were confirmed to be RAG2 KO. The thymus and spleen sizes of RAG2 KO pigs were significantly smaller than those of wild-type (WT) pigs. Hematoxylin-eosin staining results revealed that the thymus and spleen tissue structures of RAG2 KO pigs were disorganized and lacked the characteristic structures, indicating that RAG2 KO leads to dysplasia of the thymus and spleen. Hematological analysis demonstrated that the total number of white blood cells and lymphocytes in the circulation of RAG2 KO pigs was significantly lower, while the number of eosinophils was higher. Flow cytometry results indicated that the proportions of mature T and B lymphocytes were significantly reduced compared to WT pigs. These findings successfully verified the immunodeficiency phenotype of RAG2 KO pigs. This study may provide experimental animals for the development of tumor models and humanized animals.
Collapse
Affiliation(s)
- Jing Wang
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming 650201, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming 650201, China
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Feiyan Zhu
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming 650201, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming 650201, China
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Deling Jiao
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming 650201, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming 650201, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Chang Yang
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming 650201, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming 650201, China
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Junqi Wang
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming 650201, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming 650201, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Fengchong Wang
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming 650201, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming 650201, China
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Heng Zhao
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming 650201, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming 650201, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Hong-Jiang Wei
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming 650201, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming 650201, China
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Hong-Ye Zhao
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming 650201, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming 650201, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| |
Collapse
|
4
|
Zheng X, Huang C, Lin Y, Han B, Chen Y, Li C, Li J, Ding Y, Song X, Wang W, Liang W, Wu J, Wu J, Gao J, Wei C, Zhang X, Tu Z, Yan S. Generation of inactivated IL2RG and RAG1 monkeys with severe combined immunodeficiency using base editing. Signal Transduct Target Ther 2023; 8:327. [PMID: 37661226 PMCID: PMC10475462 DOI: 10.1038/s41392-023-01544-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 06/03/2023] [Accepted: 06/16/2023] [Indexed: 09/05/2023] Open
Abstract
Severe combined immunodeficiency (SCID) encompasses a range of inherited disorders that lead to a profound deterioration of the immune system. Among the pivotal genes associated with SCID, RAG1 and IL2RG play crucial roles. IL2RG is essential for the development, differentiation, and functioning of T, B, and NK cells, while RAG1 critically contributes to adaptive immunity by facilitating V(D)J recombination during the maturation of lymphocytes. Animal models carrying mutations in these genes exhibit notable deficiencies in their immune systems. Non-human primates (NHPs) are exceptionally well-suited models for biomedical research due to their genetic and physiological similarities to humans. Cytosine base editors (CBEs) serve as powerful tools for precisely and effectively modifying single-base mutations in the genome. Their successful implementation has been demonstrated in human cells, mice, and crop species. This study outlines the creation of an immunodeficient monkey model by deactivating both the IL2RG and RAG1 genes using the CBE4max system. The base-edited monkeys exhibited a severely compromised immune system characterized by lymphopenia, atrophy of lymphoid organs, and a deficiency of mature T cells. Furthermore, these base-edited monkeys were capable of hosting and supporting the growth of human breast cancer cells, leading to tumor formation. In summary, we have successfully developed an immunodeficient monkey model with the ability to foster tumor growth using the CBE4max system. These immunodeficiency monkeys show tremendous potential as valuable tools for advancing biomedical and translational research.
Collapse
Affiliation(s)
- Xiao Zheng
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, 510632, Guangzhou, China
- Department of Pathophysiology, School of Medicine, Jinan University, 510632, Guangzhou, China
| | - Chunhui Huang
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, 510632, Guangzhou, China
- Department of Pathophysiology, School of Medicine, Jinan University, 510632, Guangzhou, China
| | - Yingqi Lin
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, 510632, Guangzhou, China
- Department of Pathophysiology, School of Medicine, Jinan University, 510632, Guangzhou, China
| | - Bofeng Han
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, 510632, Guangzhou, China
| | - Yizhi Chen
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, 510632, Guangzhou, China
| | - Caijuan Li
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, 510632, Guangzhou, China
- Department of Pathophysiology, School of Medicine, Jinan University, 510632, Guangzhou, China
| | - Jiawei Li
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, 510632, Guangzhou, China
- Department of Pathophysiology, School of Medicine, Jinan University, 510632, Guangzhou, China
| | - Yongyan Ding
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, 510632, Guangzhou, China
| | - Xichen Song
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, 510632, Guangzhou, China
| | - Wei Wang
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, 510632, Guangzhou, China
| | - Weien Liang
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, 510632, Guangzhou, China
| | - Jianhao Wu
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, 510632, Guangzhou, China
| | - Jiaxi Wu
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, 510632, Guangzhou, China
| | - Jiale Gao
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, 510632, Guangzhou, China
| | - Chengxi Wei
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, 510632, Guangzhou, China
| | - Xudong Zhang
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, 510632, Guangzhou, China
| | - Zhuchi Tu
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, 510632, Guangzhou, China.
| | - Sen Yan
- Guangdong Key Laboratory of Non-human Primate Research, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, 510632, Guangzhou, China.
- Department of Pathophysiology, School of Medicine, Jinan University, 510632, Guangzhou, China.
| |
Collapse
|
5
|
Wang L, Piao Y, Guo F, Wei J, Chen Y, Dai X, Zhang X. Current progress of pig models for liver cancer research. Biomed Pharmacother 2023; 165:115256. [PMID: 37536038 DOI: 10.1016/j.biopha.2023.115256] [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: 05/17/2023] [Revised: 07/26/2023] [Accepted: 07/28/2023] [Indexed: 08/05/2023] Open
Abstract
Preclinical trials play critical roles in assessing the safety and efficiency of novel therapeutic strategies for human diseases including live cancer. However, most therapeutic strategies that were proved to be effective in preclinical cancer models failed in human clinical trials due to the lack of appropriate disease animal models. Therefore, it is of importance and urgent to develop a precise animal model for preclinical cancer research. Liver cancer is one of the most frequently diagnosed cancers with low 5-year survival rate. Recently, porcine attracted increasing attentions as animal model in biomedical research. Porcine liver cancer model may provide a promising platform for biomedical research due to their similarities to human being in body size, anatomical characteristics, physiology and pathophysiology. In this review, we comprehensively summarized and discussed the advantages and disadvantages, rationale, current status and progress of pig models for liver cancer research.
Collapse
Affiliation(s)
- Luyao Wang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital of Jilin University, Changchun, China; National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital of Jilin University, Changchun, China
| | - Yuexian Piao
- Invasive Technology Nursing Platform, First Hospital of Jilin University, Changchun, China
| | - Fucheng Guo
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital of Jilin University, Changchun, China; National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital of Jilin University, Changchun, China
| | - Jiarui Wei
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital of Jilin University, Changchun, China; National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital of Jilin University, Changchun, China
| | - Yurong Chen
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital of Jilin University, Changchun, China; National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital of Jilin University, Changchun, China
| | - Xiangpeng Dai
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital of Jilin University, Changchun, China; National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital of Jilin University, Changchun, China.
| | - Xiaoling Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital of Jilin University, Changchun, China; National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
6
|
Sarmah H, Sawada A, Hwang Y, Miura A, Shimamura Y, Tanaka J, Yamada K, Mori M. Towards human organ generation using interspecies blastocyst complementation: Challenges and perspectives for therapy. Front Cell Dev Biol 2023; 11:1070560. [PMID: 36743411 PMCID: PMC9893295 DOI: 10.3389/fcell.2023.1070560] [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: 10/15/2022] [Accepted: 01/05/2023] [Indexed: 01/20/2023] Open
Abstract
Millions of people suffer from end-stage refractory diseases. The ideal treatment option for terminally ill patients is organ transplantation. However, donor organs are in absolute shortage, and sadly, most patients die while waiting for a donor organ. To date, no technology has achieved long-term sustainable patient-derived organ generation. In this regard, emerging technologies of chimeric human organ production via blastocyst complementation (BC) holds great promise. To take human organ generation via BC and transplantation to the next step, we reviewed current emerging organ generation technologies and the associated efficiency of chimera formation in human cells from the standpoint of developmental biology.
Collapse
Affiliation(s)
- Hemanta Sarmah
- Department of Medicine, Columbia Center for Human Development, Columbia University Medical Center, New York, NY, United States
| | - Anri Sawada
- Department of Medicine, Columbia Center for Human Development, Columbia University Medical Center, New York, NY, United States
| | - Youngmin Hwang
- Department of Medicine, Columbia Center for Human Development, Columbia University Medical Center, New York, NY, United States
| | - Akihiro Miura
- Department of Medicine, Columbia Center for Human Development, Columbia University Medical Center, New York, NY, United States
| | - Yuko Shimamura
- Department of Medicine, Columbia Center for Human Development, Columbia University Medical Center, New York, NY, United States
| | - Junichi Tanaka
- Department of Medicine, Columbia Center for Human Development, Columbia University Medical Center, New York, NY, United States
| | - Kazuhiko Yamada
- Department of Surgery, Johns Hopkins University, Baltimore, MD, United States
| | - Munemasa Mori
- Department of Medicine, Columbia Center for Human Development, Columbia University Medical Center, New York, NY, United States
| |
Collapse
|
7
|
Sper RB, Proctor J, Lascina O, Guo L, Polkoff K, Kaeser T, Simpson S, Borst L, Gleason K, Zhang X, Collins B, Murphy Y, Platt JL, Piedrahita JA. Allogeneic and xenogeneic lymphoid reconstitution in a RAG2 -/- IL2RG y/- severe combined immunodeficient pig: A preclinical model for intrauterine hematopoietic transplantation. Front Vet Sci 2022; 9:965316. [PMID: 36311661 PMCID: PMC9614384 DOI: 10.3389/fvets.2022.965316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 09/20/2022] [Indexed: 11/04/2022] Open
Abstract
Mice with severe combined immunodeficiency are commonly used as hosts of human cells. Size, longevity, and physiology, however, limit the extent to which immunodeficient mice can model human systems. To address these limitations, we generated RAG2−/−IL2RGy/− immunodeficient pigs and demonstrate successful engraftment of SLA mismatched allogeneic D42 fetal liver cells, tagged with pH2B-eGFP, and human CD34+ hematopoietic stem cells after in utero cell transplantation. Following intrauterine injection at day 42–45 of gestation, fetuses were allowed to gestate to term and analyzed postnatally for the presence of pig (allogeneic) and human (xenogeneic) B cells, T-cells and NK cells in peripheral blood and other lymphoid tissues. Engraftment of allogeneic hematopoietic cells was detected based on co-expression of pH2B-eGFP and various markers of differentiation. Analysis of spleen revealed robust generation and engraftment of pH2B-eGFP mature B cells (and IgH recombination) and mature T-cells (and TCR-β recombination), T helper (CD3+CD4+) and T cytotoxic (CD3+CD8+) cells. The thymus revealed engraftment of pH2B-eGFP double negative precursors (CD4−CD8−) as well as double positive (CD4+, CD8+) precursors and single positive T-cells. After intrauterine administration of human CD34+ hematopoietic stem cells, analysis of peripheral blood and lymphoid tissues revealed the presence of human T-cells (CD3+CD4+ and CD3+CD8+) but no detectable B cells or NK cells. The frequency of human CD45+ cells in the circulation decreased rapidly and were undetectable within 2 weeks of age. The frequency of human CD45+ cells in the spleen also decreased rapidly, becoming undetectable at 3 weeks. In contrast, human CD45+CD3+T-cells comprised >70% of cells in the pig thymus at birth and persisted at the same frequency at 3 weeks. Most human CD3+ cells in the pig's thymus expressed CD4 or CD8, but few cells were double positive (CD4+ CD8+). In addition, human CD3+ cells in the pig thymus contained human T-cell excision circles (TREC), suggesting de novo development. Our data shows that the pig thymus provides a microenvironment conducive to engraftment, survival and development of human T-cells and provide evidence that the developing T-cell compartment can be populated to a significant extent by human cells in large animals.
Collapse
Affiliation(s)
- Renan B. Sper
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States,Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Jessica Proctor
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States,Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Odessa Lascina
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Ling Guo
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States,Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Kathryn Polkoff
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States,Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Tobias Kaeser
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States,Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Sean Simpson
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States,Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Luke Borst
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States,Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Katherine Gleason
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States,Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Xia Zhang
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States,Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Bruce Collins
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States,Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Yanet Murphy
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Jeffrey L. Platt
- Department of Surgery and Microbiology and Immunology, University of Michigan Health System, Ann Arbor, MI, United States
| | - Jorge A. Piedrahita
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, United States,Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States,*Correspondence: Jorge A. Piedrahita
| |
Collapse
|
8
|
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
|
9
|
Zhao H, Ye W, Guo J, Wang J, Jiao D, Xu K, Yang C, Chen S, Jamal MA, Bai Z, Wei T, Cai J, Nguyen TD, Qing Y, Cheng W, Jia B, Li H, Zhao HY, Chen Q, Wei HJ. Development of RAG2-/-IL2Rγ-/Y immune deficient FAH-knockout miniature pig. Front Immunol 2022; 13:950194. [PMID: 36032112 PMCID: PMC9400017 DOI: 10.3389/fimmu.2022.950194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
Human hepatocyte transplantation for liver disease treatment have been hampered by the lack of quality human hepatocytes. Pigs with their large body size, longevity and physiological similarities with human are appropriate animal models for the in vivo expansion of human hepatocytes. Here we report on the generation of RAG2-/-IL2Rγ-/YFAH-/- (RGFKO) pigs via CRISPR/Cas9 system and somatic cell nuclear transfer. We showed that thymic and splenic development in RGFKO pigs was impaired. V(D)J recombination processes were also inactivated. Consequently, RGFKO pigs had significantly reduced numbers of porcine T, B and NK cells. Moreover, due to the loss of FAH, porcine hepatocytes continuously undergo apoptosis and consequently suffer hepatic damage. Thus, RGFKO pigs are both immune deficient and constantly suffer liver injury in the absence of NTBC supplementation. These results suggest that RGFKO pigs have the potential to be engrafted with human hepatocytes without immune rejection, thereby allowing for large scale expansion of human hepatocytes.
Collapse
Affiliation(s)
- Heng Zhao
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Weijian Ye
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jianxiong Guo
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming, China
| | - Jiaoxiang Wang
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Deling Jiao
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming, China
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Kaixiang Xu
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming, China
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Chang Yang
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming, China
| | - Shuhan Chen
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | | | - Zhongbin Bai
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Taiyun Wei
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming, China
| | - Jie Cai
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming, China
| | - Tien Dat Nguyen
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming, China
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Yubo Qing
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Wenmin Cheng
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming, China
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Baoyu Jia
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Honghui Li
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming, China
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Hong-Ye Zhao
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming, China
- *Correspondence: Hong-Jiang Wei, ; Qingfeng Chen, ; Hong-Ye Zhao,
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- *Correspondence: Hong-Jiang Wei, ; Qingfeng Chen, ; Hong-Ye Zhao,
| | - Hong-Jiang Wei
- Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Yunnan Agricultural University, Kunming, China
- Yunnan Province Xenotransplantation Research Engineering Centre, Yunnan Agricultural University, Kunming, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- *Correspondence: Hong-Jiang Wei, ; Qingfeng Chen, ; Hong-Ye Zhao,
| |
Collapse
|
10
|
Souto EP, Dobrolecki LE, Villanueva H, Sikora AG, Lewis MT. In Vivo Modeling of Human Breast Cancer Using Cell Line and Patient-Derived Xenografts. J Mammary Gland Biol Neoplasia 2022; 27:211-230. [PMID: 35697909 PMCID: PMC9433358 DOI: 10.1007/s10911-022-09520-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 05/19/2022] [Indexed: 11/24/2022] Open
Abstract
Historically, human breast cancer has been modeled largely in vitro using long-established cell lines primarily in two-dimensional culture, but also in three-dimensional cultures of varying cellular and molecular complexities. A subset of cell line models has also been used in vivo as cell line-derived xenografts (CDX). While outstanding for conducting detailed molecular analysis of regulatory mechanisms that may function in vivo, results of drug response studies using long-established cell lines have largely failed to translate clinically. In an attempt to address this shortcoming, many laboratories have succeeded in developing clinically annotated patient-derived xenograft (PDX) models of human cancers, including breast, in a variety of host systems. While immunocompromised mice are the predominant host, the immunocompromised rat and pig, zebrafish, as well as the chicken egg chorioallantoic membrane (CAM) have also emerged as potential host platforms to help address perceived shortcomings of immunocompromised mice. With any modeling platform, the two main issues to be resolved are criteria for "credentialing" the models as valid models to represent human cancer, and utility with respect to the ability to generate clinically relevant translational research data. Such data are beginning to emerge, particularly with the activities of PDX consortia such as the NCI PDXNet Program, EuroPDX, and the International Breast Cancer Consortium, as well as a host of pharmaceutical companies and contract research organizations (CRO). This review focuses primarily on these important aspects of PDX-related research, with a focus on breast cancer.
Collapse
Affiliation(s)
- Eric P Souto
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Lacey E Dobrolecki
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Hugo Villanueva
- Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Andrew G Sikora
- Department of Head and Neck Surgery, Division of Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Michael T Lewis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA.
- Departments of Molecular and Cellular Biology and Radiology, Baylor College of Medicine, Houston, TX, 77030, USA.
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.
- Baylor College of Medicine, One Baylor Plaza, BCM-600; Room N1210, Houston, TX, 77030, USA.
| |
Collapse
|
11
|
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
|
12
|
Zbinden A, Canté-Barrett K, Pike-Overzet K, Staal FJT. Stem Cell-Based Disease Models for Inborn Errors of Immunity. Cells 2021; 11:cells11010108. [PMID: 35011669 PMCID: PMC8750661 DOI: 10.3390/cells11010108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/23/2021] [Accepted: 12/29/2021] [Indexed: 11/24/2022] Open
Abstract
The intrinsic capacity of human hematopoietic stem cells (hHSCs) to reconstitute myeloid and lymphoid lineages combined with their self-renewal capacity hold enormous promises for gene therapy as a viable treatment option for a number of immune-mediated diseases, most prominently for inborn errors of immunity (IEI). The current development of such therapies relies on disease models, both in vitro and in vivo, which allow the study of human pathophysiology in great detail. Here, we discuss the current challenges with regards to developmental origin, heterogeneity and the subsequent implications for disease modeling. We review models based on induced pluripotent stem cell technology and those relaying on use of adult hHSCs. We critically review the advantages and limitations of current models for IEI both in vitro and in vivo. We conclude that existing and future stem cell-based models are necessary tools for developing next generation therapies for IEI.
Collapse
|
13
|
Lunney JK, Van Goor A, Walker KE, Hailstock T, Franklin J, Dai C. Importance of the pig as a human biomedical model. Sci Transl Med 2021; 13:eabd5758. [PMID: 34818055 DOI: 10.1126/scitranslmed.abd5758] [Citation(s) in RCA: 296] [Impact Index Per Article: 74.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Joan K Lunney
- Animal Parasitic Diseases Laboratory, BARC, NEA, ARS, USDA, Beltsville, MD 20705, USA
| | - Angelica Van Goor
- Animal Parasitic Diseases Laboratory, BARC, NEA, ARS, USDA, Beltsville, MD 20705, USA
| | - Kristen E Walker
- Animal Parasitic Diseases Laboratory, BARC, NEA, ARS, USDA, Beltsville, MD 20705, USA
| | - Taylor Hailstock
- Animal Parasitic Diseases Laboratory, BARC, NEA, ARS, USDA, Beltsville, MD 20705, USA
| | - Jasmine Franklin
- Animal Parasitic Diseases Laboratory, BARC, NEA, ARS, USDA, Beltsville, MD 20705, USA
| | - Chaohui Dai
- Animal Parasitic Diseases Laboratory, BARC, NEA, ARS, USDA, Beltsville, MD 20705, USA.,College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China
| |
Collapse
|
14
|
Li Y, Adur MK, Wang W, Schultz RB, Hale B, Wierson W, Charley SE, McGrail M, Essner J, Tuggle CK, Ross JW. Effect of ARTEMIS (DCLRE1C) deficiency and microinjection timing on editing efficiency during somatic cell nuclear transfer and in vitro fertilization using the CRISPR/Cas9 system. Theriogenology 2021; 170:107-116. [PMID: 34004455 PMCID: PMC8243557 DOI: 10.1016/j.theriogenology.2021.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/10/2021] [Accepted: 04/14/2021] [Indexed: 01/17/2023]
Abstract
The ability to efficiently introduce site-specific genetic modifications to the mammalian genome has been dramatically improved with the use of the CRISPR/Cas9 system. CRISPR/Cas9 is a powerful tool used to generate genetic modifications by causing double-strand breaks (DSBs) in DNA. Artemis (ART; also known as DCLRE1C), is a nuclear protein and is essential for DSB end joining in DNA repair via the canonical non-homologous end joining (c-NHEJ) pathway. In this work, we tested whether ART deficiency affects DNA repair following CRISPR/Cas9 induced DSBs in somatic cells. We also demonstrated the effect of microinjection timing on embryo developmental ability and gene targeting efficiency of CRISPR/Cas9 system to disrupt the interleukin 2 receptor subunit gamma (IL2RG) locus using porcine in vitro fertilization (IVF) and somatic cell nuclear transfer (SCNT) derived embryos. In comparison to non-injected controls, CRISPR/Cas9 injection of IVF derived zygotes at 4 h and 8 h after fertilization did not impact cleavage and blastocyst rate. Gene modification rate was observed to be higher, 53.3% (9/16) in blastocysts injected 4 h post-fertilization as compared to 11.1% (1/9) in blastocysts injected 8 h post-fertilization. Microinjection 8 h after chemical activation of SCNT derived embryos decreased blastocyst development rate compared to non-injected controls but showed a higher gene modification efficiency of 66.7% as compared to 25% in the 4 h post-activation injection group. Furthermore, we observed that male ART-/- and ART+/- porcine fetal fibroblast (pFF) cells showed lower modification rates (2.5% and 1.9%, respectively) as compared to the ART intact cell line (8.3%). Interestingly, the female ART-/- and ART+/- pFF cells had modification rates (4.2% and 10.1%, respectively) similar to those seen in the ART intact cells. This study demonstrates the complex effect of various parameters such as microinjection timing and ART deficiency on gene editing efficiency in in vitro derived porcine embryos.
Collapse
Affiliation(s)
- Yunsheng Li
- Department of Animal Science, Iowa State University, Ames, Iowa, United States
| | - Malavika K. Adur
- Department of Animal Science, Iowa State University, Ames, Iowa, United States
| | - Wei Wang
- Department of Animal Science, Iowa State University, Ames, Iowa, United States
| | - R. Blythe Schultz
- Department of Animal Science, Iowa State University, Ames, Iowa, United States
| | - Benjamin Hale
- Department of Animal Science, Iowa State University, Ames, Iowa, United States
| | - Wesley Wierson
- Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, United States
| | - Sara E. Charley
- Department of Animal Science, Iowa State University, Ames, Iowa, United States
| | - Maura McGrail
- Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, United States
| | - Jeffrey Essner
- Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, United States
| | | | - Jason W. Ross
- Department of Animal Science, Iowa State University, Ames, Iowa, United States
| |
Collapse
|
15
|
Kim YY, Kim JS, Che JH, Ku SY, Kang BC, Yun JW. Comparison of Genetically Engineered Immunodeficient Animal Models for Nonclinical Testing of Stem Cell Therapies. Pharmaceutics 2021; 13:130. [PMID: 33498509 PMCID: PMC7909568 DOI: 10.3390/pharmaceutics13020130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/12/2021] [Accepted: 01/18/2021] [Indexed: 12/23/2022] Open
Abstract
For the recovery or replacement of dysfunctional cells and tissue-the goal of stem cell research-successful engraftment of transplanted cells and tissues are essential events. The event is largely dependent on the immune rejection of the recipient; therefore, the immunogenic evaluation of candidate cells or tissues in immunodeficient animals is important. Understanding the immunodeficient system can provide insights into the generation and use of immunodeficient animal models, presenting a unique system to explore the capabilities of the innate immune system. In this review, we summarize various immunodeficient animal model systems with different target genes as valuable tools for biomedical research. There have been numerous immunodeficient models developed by different gene defects, resulting in many different features in phenotype. More important, mice, rats, and other large animals exhibit very different immunological and physiological features in tissue and organs, including genetic background and a representation of human disease conditions. Therefore, the findings from this review may guide researchers to select the most appropriate immunodeficient strain, target gene, and animal species based on the research type, mutant gene effects, and similarity to human immunological features for stem cell research.
Collapse
Affiliation(s)
- Yoon-Young Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Korea; (Y.-Y.K.); (S.-Y.K.)
| | - Jin-Soo Kim
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon 14662, Korea;
| | - Jeong-Hwan Che
- Biomedical Center for Animal Resource and Development, Seoul National University College of Medicine, Seoul 03080, Korea;
| | - Seung-Yup Ku
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Korea; (Y.-Y.K.); (S.-Y.K.)
| | - Byeong-Cheol Kang
- Graduate School of Translational Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Jun-Won Yun
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon 14662, Korea;
| |
Collapse
|
16
|
Kobayashi E. Viral infections of pigs used for medical education. A Japanese experience. Acta Cir Bras 2020; 35:e202000808. [PMID: 32965305 PMCID: PMC7518223 DOI: 10.1590/s0102-865020200080000008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/11/2020] [Indexed: 08/30/2023] Open
Abstract
Infectious viruses pose a threat to all living organisms, including humans, and can cause significant morbidity. Previous experience with pigs in medical education and research, rather than in domestic control settings, has led to a unique perspective on viral infections in swine. In this article, common porcine infectious diseases have been listed, based mainly on the authors' experience thus far. For example, young domestic pigs that were used in surgical training and infected with hepatitis E were subjected to quarantine and isolation treatment, and attempts were made to develop a DNA vaccine for swine influenza arising from swine-to-human transmission. More recent research has focused on preventing infection by the African swine virus, a current threat. We hope that this article of porcine infectious diseases identified at the School of Medicine will help develop a breakthrough with regard to coronavirus disease.
Collapse
|
17
|
Boettcher AN, Cino-Ozuna AG, Solanki Y, Wiarda JE, Putz E, Owens JL, Crane SA, Ahrens AP, Loving CL, Cunnick JE, Rowland RRR, Charley SE, Dekkers JCM, Tuggle CK. CD3ε + Cells in Pigs With Severe Combined Immunodeficiency Due to Defects in ARTEMIS. Front Immunol 2020; 11:510. [PMID: 32296428 PMCID: PMC7136459 DOI: 10.3389/fimmu.2020.00510] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 03/05/2020] [Indexed: 01/24/2023] Open
Abstract
Severe combined immunodeficiency (SCID) is described as the lack of functional T and B cells. In some cases, mutant genes encoding proteins involved in the process of VDJ recombination retain partial activity and are classified as hypomorphs. Hypomorphic activity in the products from these genes can function in the development of T and B cells and is referred to as a leaky phenotype in patients and animals diagnosed with SCID. We previously described two natural, single nucleotide variants in ARTEMIS (DCLR1EC) in a line of Yorkshire pigs that resulted in SCID. One allele contains a splice site mutation within intron 8 of the ARTEMIS gene (ART16), while the other mutation is within exon 10 that results in a premature stop codon (ART12). While initially characterized as SCID and lacking normal levels of circulating lymphoid cells, low levels of CD3ε+ cells can be detected in most SCID animals. Upon further assessment, we found that ART16/16, and ART12/12 SCID pigs had abnormally small populations of CD3ε+ cells, but not CD79α+ cells, in circulation and lymph nodes. Newborn pigs (0 days of age) had CD3ε+ cells within lymph nodes prior to any environmental exposure. CD3ε+ cells in SCID pigs appeared to have a skewed CD4α+CD8α+CD8β− T helper memory phenotype. Additionally, in some pigs, rearranged VDJ joints were detected in lymph node cells as probed by PCR amplification of TCRδ V5 and J1 genomic loci, as well as TCRβ V20 and J1.1, providing molecular evidence of residual Artemis activity. We additionally confirmed that TCRα and TCRδ constant region transcripts were expressed in the thymic and lymph node tissues of SCID pigs; although the expression pattern was abnormal compared to carrier animals. The leaky phenotype is important to characterize, as SCID pigs are an important tool for biomedical research and this additional phenotype may need to be considered. The pig model also provides a relevant model for hypomorphic human SCID patients.
Collapse
Affiliation(s)
- Adeline N Boettcher
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - A Giselle Cino-Ozuna
- Veterinary Diagnostic Laboratory, Kansas State University, Manhattan, KS, United States
| | - Yash Solanki
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Jayne E Wiarda
- Food Safety and Enteric Pathogen Unit, National Animal Disease Center, Agricultural Research Service, U.S. Department of Agriculture, Ames, IA, United States.,Immunobiology Graduate Program, College of Veterinary Medicine, Iowa State University, Ames, IA, United States.,Agricultural Research Service Participation Program, Oak Ridge Institute for Science and Education, Oak Ridge, TN, United States
| | - Ellie Putz
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Jeana L Owens
- Veterinary Diagnostic Laboratory, Kansas State University, Manhattan, KS, United States
| | - Sara A Crane
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Amanda P Ahrens
- Laboratory Animal Research, Iowa State University, Ames, IA, United States
| | - Crystal L Loving
- Food Safety and Enteric Pathogen Unit, National Animal Disease Center, Agricultural Research Service, U.S. Department of Agriculture, Ames, IA, United States
| | - Joan E Cunnick
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Raymond R R Rowland
- Diagnostic Medicine and Pathobiology Department, Kansas State University, Manhattan, KS, United States
| | - Sara E Charley
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Jack C M Dekkers
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | | |
Collapse
|