1
|
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
|
2
|
Widmer KM, Rahic-Seggerman F, Forster A, Ahrens-Kress A, Sauer M, Mooyottu S, Vinithakumari A, Dunkerson-Kurzhumov A, Sponseller B, Kiupel M, Schmitz-Esser S, Tuggle CK. EFFECT OF GENOTYPE AND AGE ON A DEFINED MICROBIOTA IN GNOTOBIOTIC SCID PIGLETS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.03.611011. [PMID: 39282343 PMCID: PMC11398413 DOI: 10.1101/2024.09.03.611011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
Severe combined immunodeficient (SCID) individuals lack functional T and B lymphocytes, leading to a deficient adaptive immune system. SCID pigs are a unique large animal biomedical model as they possess many similarities to humans, allowing for the collection of translatable data in regenerative medicine, cancer, and other biomedical research topics. While many studies suggest early gut microbiota development is necessary for developing the intestinal barrier and immune system, these animals are often cesarian section derived, leaving them uncolonized for normal intestinal microflora. The hypothesis was that an increase in complexity of microbiota inoculum will allow for more stability in the composition of the gut microbiota of SCID piglets. This was tested across multiple litters of SCID piglets with three different defined microbiota consortium (2-strain, 6-strain, 7-strain). All piglets received their designated defined microbiota by oral gavage immediately after birth and again 24 hours later. There was no effect of SCID genotype on the composition of the gut microbiota, but there was a significant effect due to piglet age. Additionally, all three defined microbiota consortia were deemed safe to use in SCID piglets, and the 7-strain microbiota was the most stable over time. Based on these results, the 7-strain defined microbiota will be added to the SCID pig husbandry protocol, allowing for a more reproducible model.
Collapse
Affiliation(s)
- Katherine M Widmer
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA
| | | | - Ahlea Forster
- Department of Animal Science, Iowa State University, Ames, IA 50011, USA
| | | | - Mary Sauer
- Laboratory Animal Resources, Iowa State University, Ames, IA 50011, USA
| | - Shankumar Mooyottu
- Department of Veterinary Pathology, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA
- Department of Pathobiology, Auburn University, Auburn, AL, 36849, USA
| | - Akhil Vinithakumari
- Department of Veterinary Pathology, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA
| | - Aaron Dunkerson-Kurzhumov
- Department of Veterinary Pathology, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA
| | - Brett Sponseller
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA
| | - Matti Kiupel
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, 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
|
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
|
5
|
Teper D, White FF, Wang N. The Dynamic Transcription Activator-Like Effector Family of Xanthomonas. PHYTOPATHOLOGY 2023; 113:651-666. [PMID: 36449529 DOI: 10.1094/phyto-10-22-0365-kd] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Transcription activator-like effectors (TALEs) are bacterial proteins that are injected into the eukaryotic nucleus to act as transcriptional factors and function as key virulence factors of the phytopathogen Xanthomonas. TALEs are translocated into plant host cells via the type III secretion system and induce the expression of host susceptibility (S) genes to facilitate disease. The unique modular DNA binding domains of TALEs comprise an array of nearly identical direct repeats that enable binding to DNA targets based on the recognition of a single nucleotide target per repeat. The very nature of TALE structure and function permits the proliferation of TALE genes and evolutionary adaptations in the host to counter TALE function, making the TALE-host interaction the most dynamic story in effector biology. The TALE genes appear to be a relatively young effector gene family, with a presence in all virulent members of some species and absent in others. Genome sequencing has revealed many TALE genes throughout the xanthomonads, and relatively few have been associated with a cognate S gene. Several species, including Xanthomonas oryzae pv. oryzae and X. citri pv. citri, have near absolute requirement for TALE gene function, while the genes appear to be just now entering the disease interactions with new fitness contributions to the pathogens of tomato and pepper among others. Deciphering the simple and effective DNA binding mechanism also has led to the development of DNA manipulation tools in fields of gene editing and transgenic research. In the three decades since their discovery, TALE research remains at the forefront of the study of bacterial evolution, plant-pathogen interactions, and synthetic biology. We also discuss critical questions that remain to be addressed regarding TALEs.
Collapse
Affiliation(s)
- Doron Teper
- Department of Plant Pathology and Weed Research, Agricultural Research Organization, Volcani Institute, Rishon LeZion, Israel
| | - Frank F White
- Department of Plant Pathology, Institute of Food and Agricultural Sciences (IFAS), University of Florida, Gainesville, FL, U.S.A
| | - Nian Wang
- Citrus Research and Education Center, Department of Microbiology and Cell Science, IFAS, University of Florida, Lake Alfred, FL, U.S.A
| |
Collapse
|
6
|
Hu D, Li X, Li J, Tong P, Li Z, Lin G, Sun Y, Wang J. The preclinical and clinical progress of cell sheet engineering in regenerative medicine. Stem Cell Res Ther 2023; 14:112. [PMID: 37106373 PMCID: PMC10136407 DOI: 10.1186/s13287-023-03340-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Cell therapy is an accessible method for curing damaged organs or tissues. Yet, this approach is limited by the delivery efficiency of cell suspension injection. Over recent years, biological scaffolds have emerged as carriers of delivering therapeutic cells to the target sites. Although they can be regarded as revolutionary research output and promote the development of tissue engineering, the defect of biological scaffolds in repairing cell-dense tissues is apparent. Cell sheet engineering (CSE) is a novel technique that supports enzyme-free cell detachment in the shape of a sheet-like structure. Compared with the traditional method of enzymatic digestion, products harvested by this technique retain extracellular matrix (ECM) secreted by cells as well as cell-matrix and intercellular junctions established during in vitro culture. Herein, we discussed the current status and recent progress of CSE in basic research and clinical application by reviewing relevant articles that have been published, hoping to provide a reference for the development of CSE in the field of stem cells and regenerative medicine.
Collapse
Affiliation(s)
- Danping Hu
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410008, China
- HANGZHOU CHEXMED TECHNOLOGY CO., LTD, Hangzhou, 310000, China
| | - Xinyu Li
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410008, China
| | - Jie Li
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410008, China
| | - Pei Tong
- Hospital of Hunan Guangxiu, Medical College of Hunan Normal University, Hunan Normal University, Changsha, 410008, China
| | - Zhe Li
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410008, China
| | - Ge Lin
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410008, China
- National Engineering and Research Center of Human Stem Cells, Changsha, 410008, China
- Key Laboratory of Stem Cells and Reproductive Engineering, Ministry of Health, Changsha, 410008, China
| | - Yi Sun
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, 410008, China.
- National Engineering and Research Center of Human Stem Cells, Changsha, 410008, China.
- Key Laboratory of Stem Cells and Reproductive Engineering, Ministry of Health, Changsha, 410008, China.
| | - Juan Wang
- Shanghai Biomass Pharmaceutical Product Evaluation Professional Public Service Platform, Center for Pharmacological Evaluation and Research, China State Institute of Pharmaceutical Industry, Shanghai, 200437, China.
| |
Collapse
|
7
|
Chen PR, Uh K, Monarch K, Spate LD, Reese ED, Prather RS, Lee K. Inactivation of growth differentiation factor 9 blocks folliculogenesis in pigs†. Biol Reprod 2023; 108:611-618. [PMID: 36648449 PMCID: PMC10106843 DOI: 10.1093/biolre/ioad005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 12/20/2022] [Accepted: 01/10/2023] [Indexed: 01/18/2023] Open
Abstract
Growth differentiation factor 9 (GDF9) is a secreted protein belonging to the transforming growth factor beta superfamily and has been well characterized for its role during folliculogenesis in the ovary. Although previous studies in mice and sheep have shown that mutations in GDF9 disrupt follicular progression, the exact role of GDF9 in pigs has yet to be elucidated. The objective of this study was to understand the role of GDF9 in ovarian function by rapidly generating GDF9 knockout (GDF9-/-) pigs by using the CRISPR/Cas9 system. Three single-guide RNAs designed to disrupt porcine GDF9 were injected with Cas9 mRNA into zygotes, and blastocyst-stage embryos were transferred into surrogates. One pregnancy was sacrificed on day 100 of gestation to investigate the role of GDF9 during oogenesis. Four female fetuses were recovered with one predicted to be GDF9-/- and the others with in-frame mutations. All four had fully formed oocytes within primordial follicles, confirming that knockout of GDF9 does not disrupt oogenesis. Four GDF9 mutant gilts were generated and were grown past puberty. One gilt was predicted to completely lack functional GDF9 (GDF9-/-), and the gilt never demonstrated standing estrus and had a severely underdeveloped reproductive tract with large ovarian cysts. Further examination revealed that the follicles from the GDF9-/- gilt did not progress past preantral stages, and the uterine vasculature was less extensive than the control pigs. By using the CRISPR/Cas9 system, we demonstrated that GDF9 is a critical growth factor for proper ovarian development and function in pigs.
Collapse
Affiliation(s)
- Paula R Chen
- United States Department of Agriculture—Agricultural Research Service, Plant Genetics Research Unit, Columbia, MO, USA
| | - Kyungjun Uh
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Kaylynn Monarch
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Lee D Spate
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
- National Swine Resource and Research Center, University of Missouri, Columbia, MO, USA
| | - Emily D Reese
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Randall S Prather
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
- National Swine Resource and Research Center, University of Missouri, Columbia, MO, USA
| | - Kiho Lee
- Division of Animal Sciences, University of Missouri, Columbia, MO, USA
- National Swine Resource and Research Center, University of Missouri, Columbia, MO, USA
| |
Collapse
|
8
|
Hasegawa K, Nakano K, Nagaya M, Watanabe M, Uchikura A, Matsunari H, Umeyama K, Kobayashi E, Nagashima H. Transplantation of human cells into Interleukin-2 receptor gamma gene knockout pigs under several conditions. Regen Ther 2022; 21:62-72. [PMID: 35765545 PMCID: PMC9198816 DOI: 10.1016/j.reth.2022.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/22/2022] [Accepted: 05/25/2022] [Indexed: 11/30/2022] Open
Abstract
Introduction Previously, we performed gene knockout (KO) of interleukin-2 receptor gamma (IL2RG) in porcine fetal fibroblasts using zinc finger nuclease-encoding mRNAs, subsequently generating IL2RG KO pigs using these cells through somatic cell nuclear transfer. The IL2RG KO pigs lacked a thymus and were deficient in T lymphocytes and natural killer cells, similar to human X-linked severe combined immunodeficiency (SCID) patients. The present study aimed to evaluate whether pigs can support the growth of xenografted human cells and have the potential to be an effective animal model. Methods The IL2RG XKOY pigs used in this study were obtained by mating IL2RG XKOX females with wild-type boars. This permitted the routine production of IL2RG KO pigs via natural breeding without complicated somatic cell cloning procedures; therefore, a sufficient number of pigs could be prepared. We transplanted human HeLa S3 cells expressing the tandem dimer tomato into the ears and pancreas of IL2RG KO pigs. Additionally, a newly developed method for the aseptic rearing of SCID pigs was used in case of necessity. Results Tumors from the transplanted cells quickly developed in all pigs and were verified by histology and immunohistochemistry. We also transplanted these cells into the pancreas of designated pathogen-free pigs housed in novel biocontainment facilities, and large tumors were confirmed. Conclusions IL2RG KO pigs have the potential to become useful animal models in a variety of translational biology fields. The present study aimed to evaluate whether IL2RG KO SCID-like pigs can host and support the growth of xenografted human cells under several conditions. Tumors from transplanted cells quickly developed in all pigs, as verified by histology and immunohistochemistry. IL2RG KO pigs have the potential to become extremely useful animal models in a variety of translational biology fields.
Collapse
Key Words
- DPF, designated pathogen-free
- IL, Interleukin
- IL2RG, interleukin-2 receptor gamma
- Interleukin-2 receptor gamma
- KO, knock out pigs
- NK cells, natural killer cells
- OIDP, operational immunodeficient pig
- PCR, polymerase chain reaction
- Pig
- SCID
- SCID, Severe combined immunodeficiency
- SCNT, somatic cell nuclear transfer
- SD, standard deviation
- U-iR, uterectomy-isolated rearing
- WT, wild-type pigs
- XLGD, X-linked genetic diseases
- Xenotransplantation
- ZFN, Zinc finger nuclease
- tdTomato, tandem dimer Tomato
Collapse
Affiliation(s)
- Koki Hasegawa
- Laboratory of Developmental Engineering, Department of Life Sciences, School of Agriculture, Meiji University, 1-1-1 Higashimita, Tama-ku, Kawasaki 214-8571, Japan
| | - Kazuaki Nakano
- PorMedTec Co. Ltd., 2-3227 MIta, Tama-ku, Kawasaki, Kanagawa, 214-0034, Japan.,Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki 214-8571, Japan
| | - Masaki Nagaya
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki 214-8571, Japan
| | - Masahito Watanabe
- PorMedTec Co. Ltd., 2-3227 MIta, Tama-ku, Kawasaki, Kanagawa, 214-0034, Japan.,Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki 214-8571, Japan
| | - Ayuko Uchikura
- Laboratory of Developmental Engineering, Department of Life Sciences, School of Agriculture, Meiji University, 1-1-1 Higashimita, Tama-ku, Kawasaki 214-8571, Japan.,Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki 214-8571, Japan
| | - Hitomi Matsunari
- Laboratory of Developmental Engineering, Department of Life Sciences, School of Agriculture, Meiji University, 1-1-1 Higashimita, Tama-ku, Kawasaki 214-8571, Japan.,Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki 214-8571, Japan
| | - Kazuhiro Umeyama
- PorMedTec Co. Ltd., 2-3227 MIta, Tama-ku, Kawasaki, Kanagawa, 214-0034, Japan.,Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki 214-8571, Japan
| | - Eiji Kobayashi
- Department of Kidney Regenerative Medicine, The Jikei University School of Medicine - Tokyo, Japan
| | - Hiroshi Nagashima
- Laboratory of Developmental Engineering, Department of Life Sciences, School of Agriculture, Meiji University, 1-1-1 Higashimita, Tama-ku, Kawasaki 214-8571, Japan.,Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki 214-8571, Japan
| |
Collapse
|
9
|
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
|
10
|
Faridar A, Vasquez M, Thome AD, Yin Z, Xuan H, Wang JH, Wen S, Li X, Thonhoff JR, Zhao W, Zhao H, Beers DR, Wong STC, Masdeu JC, Appel SH. Ex vivo expanded human regulatory T cells modify neuroinflammation in a preclinical model of Alzheimer's disease. Acta Neuropathol Commun 2022; 10:144. [PMID: 36180898 PMCID: PMC9524037 DOI: 10.1186/s40478-022-01447-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022] Open
Abstract
Background Regulatory T cells (Tregs) play a neuroprotective role by suppressing microglia and macrophage-mediated inflammation and modulating adaptive immune reactions. We previously documented that Treg immunomodulatory mechanisms are compromised in Alzheimer’s disease (AD). Ex vivo expansion of Tregs restores and amplifies their immunosuppressive functions in vitro. A key question is whether adoptive transfer of ex vivo expanded human Tregs can suppress neuroinflammation and amyloid pathology in a preclinical mouse model. Methods An immunodeficient mouse model of AD was generated by backcrossing the 5xFAD onto Rag2 knockout mice (5xFAD-Rag2KO). Human Tregs were expanded ex vivo for 24 days and administered to 5xFAD-Rag2KO. Changes in amyloid burden, microglia characteristics and reactive astrocytes were evaluated using ELISA and confocal microscopy. NanoString Mouse AD multiplex gene expression analysis was applied to explore the impact of ex vivo expanded Tregs on the neuroinflammation transcriptome. Results Elimination of mature B and T lymphocytes and natural killer cells in 5xFAD-Rag2KO mice was associated with upregulation of 95 inflammation genes and amplified number of reactive microglia within the dentate gyrus. Administration of ex vivo expanded Tregs reduced amyloid burden and reactive glial cells in the dentate gyrus and frontal cortex of 5xFAD-Rag2KO mice. Interrogation of inflammation gene expression documented down-regulation of pro-inflammatory cytokines (IL1A&B, IL6), complement cascade (C1qa, C1qb, C1qc, C4a/b), toll-like receptors (Tlr3, Tlr4 and Tlr7) and microglial activations markers (CD14, Tyrobp,Trem2) following Treg administration. Conclusions Ex vivo expanded Tregs with amplified immunomodulatory function, suppressed neuroinflammation and alleviated AD pathology in vivo. Our results provide preclinical evidences for Treg cell therapy as a potential treatment strategy in AD. Supplementary Information The online version contains supplementary material available at 10.1186/s40478-022-01447-z.
Collapse
Affiliation(s)
- Alireza Faridar
- Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, 6560 Fannin Street, Suite ST-802, Houston, TX, 77030, USA
| | - Matthew Vasquez
- Systems Medicine and Bioengineering Department, Houston Methodist Cancer Center, Houston, TX, USA
| | - Aaron D Thome
- Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, 6560 Fannin Street, Suite ST-802, Houston, TX, 77030, USA
| | - Zheng Yin
- Systems Medicine and Bioengineering Department, Houston Methodist Cancer Center, Houston, TX, USA
| | - Hui Xuan
- Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, 6560 Fannin Street, Suite ST-802, Houston, TX, 77030, USA
| | - Jing Hong Wang
- Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, 6560 Fannin Street, Suite ST-802, Houston, TX, 77030, USA
| | - Shixiang Wen
- Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, 6560 Fannin Street, Suite ST-802, Houston, TX, 77030, USA
| | - Xuping Li
- T. T. and W. F. Chao Center for BRAIN, Houston Methodist Hospital, Houston, TX, USA
| | - Jason R Thonhoff
- Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, 6560 Fannin Street, Suite ST-802, Houston, TX, 77030, USA
| | - Weihua Zhao
- Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, 6560 Fannin Street, Suite ST-802, Houston, TX, 77030, USA
| | - Hong Zhao
- Systems Medicine and Bioengineering Department, Houston Methodist Cancer Center, Houston, TX, USA
| | - David R Beers
- Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, 6560 Fannin Street, Suite ST-802, Houston, TX, 77030, USA
| | - Stephen T C Wong
- Systems Medicine and Bioengineering Department, Houston Methodist Cancer Center, Houston, TX, USA
| | - Joseph C Masdeu
- Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, 6560 Fannin Street, Suite ST-802, Houston, TX, 77030, USA
| | - Stanley H Appel
- Stanley H. Appel Department of Neurology, Houston Methodist Research Institute, 6560 Fannin Street, Suite ST-802, Houston, TX, 77030, USA.
| |
Collapse
|
11
|
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: 0.5] [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
|
12
|
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: 1.5] [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
|
13
|
Hou N, Du X, Wu S. Advances in pig models of human diseases. Animal Model Exp Med 2022; 5:141-152. [PMID: 35343091 PMCID: PMC9043727 DOI: 10.1002/ame2.12223] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 02/14/2022] [Accepted: 03/02/2022] [Indexed: 01/07/2023] Open
Abstract
Animal models of human diseases play a critical role in medical research. Pigs are anatomically and physiologically more like humans than are small rodents such as mice, making pigs an attractive option for modeling human diseases. Advances in recent years in genetic engineering have facilitated the rapid rise of pig models for use in studies of human disease. In the present review, we summarize the current status of pig models for human cardiovascular, metabolic, neurodegenerative, and various genetic diseases. We also discuss areas that need to be improved. Animal models of human diseases play a critical role in medical research. Advances in recent years in genetic engineering have facilitated the rapid rise of pig models for use in studies of human disease. In the present review, we summarize the current status of pig models for human cardiovascular, metabolic, neurodegenerative, various genetic diseases and xenotransplantation.
Collapse
Affiliation(s)
- Naipeng Hou
- College of Animal Science and Technology, China Agricultural University, Beijing, China.,Sanya Institute of China Agricultural University, Sanya, China
| | - Xuguang Du
- Sanya Institute of China Agricultural University, Sanya, China.,State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Sen Wu
- College of Animal Science and Technology, China Agricultural University, Beijing, China.,Sanya Institute of China Agricultural University, Sanya, China.,State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| |
Collapse
|
14
|
Chen PR, Uh K, Redel BK, Reese ED, Prather RS, Lee K. Production of Pigs From Porcine Embryos Generated in vitro. FRONTIERS IN ANIMAL SCIENCE 2022. [DOI: 10.3389/fanim.2022.826324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Generating porcine embryos in vitro is a critical process for creating genetically modified pigs as agricultural and biomedical models; however, these embryo technologies have been scarcely applied by the swine industry. Currently, the primary issue with in vitro-produced porcine embryos is low pregnancy rate after transfer and small litter size, which may be exasperated by micromanipulation procedures. Thus, in this review, we discuss improvements that have been made to the in vitro porcine embryo production system to increase the number of live piglets per pregnancy as well as abnormalities in the embryos and piglets that may arise from in vitro culture and manipulation techniques. Furthermore, we examine areas related to embryo production and transfer where improvements are warranted that will have direct applications for increasing pregnancy rate after transfer and the number of live born piglets per litter.
Collapse
|
15
|
Ren J, Yu D, Wang J, Xu K, Xu Y, Sun R, An P, Li C, Feng G, Zhang Y, Dai X, Zhao H, Wang Z, Han Z, Zhu H, Ding Y, You X, Liu X, Wu M, Luo L, Li Z, Yang YG, Hu Z, Wei HJ, Ge L, Hai T, Li W. Generation of immunodeficient pig with hereditary tyrosinemia type 1 and their preliminary application for humanized liver. Cell Biosci 2022; 12:26. [PMID: 35255981 PMCID: PMC8900390 DOI: 10.1186/s13578-022-00760-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 02/08/2022] [Indexed: 01/17/2023] Open
Abstract
Background Mice with humanized livers are important models to study drug toxicology testing, development of hepatitis virus treatments, and hepatocyte transplantation therapy. However, the huge difference between mouse and human in size and anatomy limited the application of humanized mice in investigating human diseases. Therefore, it is urgent to construct humanized livers in pigs to precisely investigate hepatocyte regeneration and human hepatocyte therapy. CRISPR/Cas9 system and somatic cell cloning technology were used to generate two pig models with FAH deficiency and exhibiting severe immunodeficiency (FAH/RAG1 and FAH/RAG1/IL2RG deficiency). Human primary hepatocytes were then successfully transplanted into the FG pig model and constructed two pigs with human liver. Results The constructed FAH/RAG1/IL2RG triple-knockout pig models were characterized by chronic liver injury and severe immunodeficiency. Importantly, the FG pigs transplanted with primary human hepatocytes produced human albumin in a time dependent manner as early as 1 week after transplantation. Furthermore, the colonization of human hepatocytes was confirmed by immunochemistry staining. Conclusions We successfully generated pig models with severe immunodeficiency that could construct human liver tissues. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-022-00760-3.
Collapse
Affiliation(s)
- Jilong Ren
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, 100101, China.,Institute of Animal Sciences (IAS), Chinese Academy of Agricultural Sciences (CAAS), Beijing, 100193, China
| | - Dawei Yu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China. .,Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, 100101, China. .,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Jing Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kai Xu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, 100101, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Yanan Xu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Renren Sun
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, First Hospital, Jilin University, Changchun, 130062, China
| | - Peipei An
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, First Hospital, Jilin University, Changchun, 130062, China
| | - Chongyang Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, 100101, China
| | - Guihai Feng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, 100101, China
| | - Ying Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xiangpeng Dai
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, First Hospital, Jilin University, Changchun, 130062, China
| | - Hongye Zhao
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China
| | - Zhengzhu Wang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, First Hospital, Jilin University, Changchun, 130062, China
| | - Zhiqiang Han
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, 100101, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Haibo Zhu
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, First Hospital, Jilin University, Changchun, 130062, China.,Center of Reproductive Medicine and Center of Prenatal Diagnosis, First Hospital, Jilin University, Changchun, 130021, China
| | - Yuchun Ding
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China.,Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing, 402460, China.,Chongqing Key Laboratory of Pig Industry Sciences, Chongqing, 402460, China.,Technical Engineering Center for the Development and Utilization of Medical Animal Resources, Chongqing, 402460, China
| | - Xiaoyan You
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China.,Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing, 402460, China.,Chongqing Key Laboratory of Pig Industry Sciences, Chongqing, 402460, China.,Technical Engineering Center for the Development and Utilization of Medical Animal Resources, Chongqing, 402460, China
| | - Xueqin Liu
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China.,Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing, 402460, China.,Chongqing Key Laboratory of Pig Industry Sciences, Chongqing, 402460, China.,Technical Engineering Center for the Development and Utilization of Medical Animal Resources, Chongqing, 402460, China
| | - Meng Wu
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China.,Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing, 402460, China.,Chongqing Key Laboratory of Pig Industry Sciences, Chongqing, 402460, China.,Technical Engineering Center for the Development and Utilization of Medical Animal Resources, Chongqing, 402460, China
| | - Lin Luo
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China.,Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing, 402460, China.,Chongqing Key Laboratory of Pig Industry Sciences, Chongqing, 402460, China.,Technical Engineering Center for the Development and Utilization of Medical Animal Resources, Chongqing, 402460, China
| | - Ziyi Li
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, First Hospital, Jilin University, Changchun, 130062, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, First Hospital, Jilin University, Changchun, 130062, China
| | - Zheng Hu
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, First Hospital, Jilin University, Changchun, 130062, China.
| | - Hong-Jiang Wei
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China.
| | - Liangpeng Ge
- Chongqing Academy of Animal Sciences, Chongqing, 402460, China. .,Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing, 402460, China. .,Chongqing Key Laboratory of Pig Industry Sciences, Chongqing, 402460, China. .,Technical Engineering Center for the Development and Utilization of Medical Animal Resources, Chongqing, 402460, China.
| | - Tang Hai
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China. .,Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, 100101, China. .,Beijing Farm Animal Research Center, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China. .,Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, 100101, China. .,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
16
|
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: 1.5] [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
|
17
|
Use of Genome Editing Techniques to Produce Transgenic Farm Animals. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1354:279-297. [PMID: 34807447 PMCID: PMC9810480 DOI: 10.1007/978-3-030-85686-1_14] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Recombinant proteins are essential for the treatment and diagnosis of clinical human ailments. The availability and biological activity of recombinant proteins is heavily influenced by production platforms. Conventional production platforms such as yeast, bacteria, and mammalian cells have biological and economical challenges. Transgenic livestock species have been explored as an alternative production platform for recombinant proteins, predominantly through milk secretion; the strategy has been demonstrated to produce large quantities of biologically active proteins. The major limitation of utilizing livestock species as bioreactors has been efforts required to alter the genome of livestock. Advancements in the genome editing field have drastically improved the ability to genetically engineer livestock species. Specifically, genome editing tools such as the CRISPR/Cas9 system have lowered efforts required to generate genetically engineered livestock, thus minimizing restrictions on the type of genetic modification in livestock. In this review, we discuss characteristics of transgenic animal bioreactors and how the use of genome editing systems enhances design and availability of the animal models.
Collapse
|
18
|
Hendricks-Wenger A, Nagai-Singer MA, Uh K, Vlaisavljevich E, Lee K, Allen IC. Employing Novel Porcine Models of Subcutaneous Pancreatic Cancer to Evaluate Oncological Therapies. Methods Mol Biol 2022; 2394:883-895. [PMID: 35094364 DOI: 10.1007/978-1-0716-1811-0_47] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Immunocompromised mice are commonly utilized to study pancreatic cancer and other malignancies. The ability to xenograft tumors in either subcutaneous or orthotopic locations provides a robust model to study diverse biological features of human malignancies. However, there is a dire need for large animal models that better recapitulate human anatomy in terms of size and physiology. These models will be critical for biomedical device development, surgical optimization, and drug discovery. Here, we describe the generation and application of immunocompromised pigs lacking RAG2 and IL2RG as a novel model for human xenograft studies. These SCID-like pigs closely resemble NOD scid gamma mice and are receptive to human tumor tissue, cell lines, and organoid xenografts. However, due to their immunocompromised nature, these immunocompromised animals require housing and maintenance under germfree conditions. In this protocol, we describe the use of these pigs in a subcutaneous tumor injection study with human PANC1 cells. The tumors demonstrate a steady, linear growth curve, reaching 1.0 cm within 30 days post injection. The model described here is focused on subcutaneous injections behind the ear. However, it is readily adaptable for other locations and additional human cell types.
Collapse
Affiliation(s)
- Alissa Hendricks-Wenger
- Graduate Program in Translational Biology, Medicine and Health, Virginia Polytechnic Institute and State University, Roanoke, VA, USA
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, USA
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Margaret A Nagai-Singer
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, USA
| | - Kyungjun Uh
- Department of Animal and Poultry Sciences, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA, USA
| | - Eli Vlaisavljevich
- Graduate Program in Translational Biology, Medicine and Health, Virginia Polytechnic Institute and State University, Roanoke, VA, USA
- Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Kiho Lee
- Department of Animal and Poultry Sciences, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA, USA
| | - Irving C Allen
- Graduate Program in Translational Biology, Medicine and Health, Virginia Polytechnic Institute and State University, Roanoke, VA, USA.
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, USA.
| |
Collapse
|
19
|
Hendricks-Wenger A, Arnold L, Gannon J, Simon A, Singh N, Sheppard H, Nagai-Singer MA, Imran KM, Lee K, Clark-Deener S, Byron C, Edwards MR, Larson MM, Rossmeisl JH, Coutermarsh-Ott SL, Eden K, Dervisis N, Klahn S, Tuohy J, Allen IC, Vlaisavljevich E. Histotripsy Ablation in Preclinical Animal Models of Cancer and Spontaneous Tumors in Veterinary Patients: A Review. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2022; 69:5-26. [PMID: 34478363 PMCID: PMC9284566 DOI: 10.1109/tuffc.2021.3110083] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
New therapeutic strategies are direly needed in the fight against cancer. Over the last decade, several tumor ablation strategies have emerged as stand-alone or combination therapies. Histotripsy is the first completely noninvasive, nonthermal, and nonionizing tumor ablation method. Histotripsy can produce consistent and rapid ablations, even near critical structures. Additional benefits include real-time image guidance, high precision, and the ability to treat tumors of any predetermined size and shape. Unfortunately, the lack of clinically and physiologically relevant preclinical cancer models is often a significant limitation with all focal tumor ablation strategies. The majority of studies testing histotripsy for cancer treatment have focused on small animal models, which have been critical in moving this field forward and will continue to be essential for providing mechanistic insight. While these small animal models have notable translational value, there are significant limitations in terms of scale and anatomical relevance. To address these limitations, a diverse range of large animal models and spontaneous tumor studies in veterinary patients have emerged to complement existing rodent models. These models and veterinary patients are excellent at providing realistic avenues for developing and testing histotripsy devices and techniques designed for future use in human patients. Here, we provide a review of animal models used in preclinical histotripsy studies and compare histotripsy ablation in these models using a series of original case reports across a broad spectrum of preclinical animal models and spontaneous tumors in veterinary patients.
Collapse
|
20
|
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: 269] [Impact Index Per Article: 89.7] [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
|
21
|
Nelson ED, Larson E, Joo DJ, Mao S, Glorioso J, Abu Rmilah A, Zhou W, Jia Y, Mounajjed T, Shi M, Bois M, Wood A, Jin F, Whitworth K, Wells K, Spate A, Samuel M, Minshew A, Walters E, Rinaldo P, Lillegard J, Johnson A, Amiot B, Hickey R, Prather R, Platt JL, Nyberg SL. Limited Expansion of Human Hepatocytes in FAH/RAG2-Deficient Swine. Tissue Eng Part A 2021; 28:150-160. [PMID: 34309416 PMCID: PMC8892989 DOI: 10.1089/ten.tea.2021.0057] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND The mammalian liver's regenerative ability has led researchers to engineer animals as incubators for expansion of human hepatocytes. The expansion properties of human hepatocytes in immunodeficient mice are well known. However, little has been reported about larger animals that are more scalable and practical for clinical purposes. Therefore, we engineered immunodeficient swine to support expansion of human hepatocytes and identify barriers to their clinical application. METHODS Immunodeficient swine were engineered by knockout of recombinase activating gene 2 (RAG2) and fumarylacetoacetate hydrolase (FAH). Immature human hepatocytes (ihHCs) were injected into fetal swine by intrauterine cell transplantation (IUCT) at day 40 of gestation. Human albumin was measured as a marker of engraftment. Cytotoxicity against ihHCs was measured in transplanted piglets and control swine. RESULTS Higher levels of human albumin were detected in cord blood of newborn FAH/RAG2-deficient (FR) pigs compared to immunocompetent controls (196.26 ng/dL vs 39.29 ng/dL, p = 0.008), indicating successful engraftment of ihHC after IUCT and adaptive immunity in the fetus. Although rare hepatocytes staining positively for human albumin were observed, levels of human albumin did not rise after birth but declined suggesting rejection of xenografted ihHCs. Cytotoxicity against ihHCs increased after birth 3.8% (95% CI: [2.1%, 5.4%], p < 0.001) and correlated inversely to declining levels of human albumin (p = 2.1 x 10-5, R2 = 0.17). Circulating numbers of T-cells and B-cells were negligible in FR pigs. However, circulating natural killer (NK) cells exerted cytotoxicity against ihHCs. NK cell activity was lower in immunodeficient piglets after IUCT than naive controls (30.4% vs 40.1% (p = 0.011, 95% CI for difference [2.7%, 16.7%]). CONCLUSION Immature human hepatocytes successfully engrafted in FR swine after IUCT. NK cells were a significant barrier to expansion of hepatocytes. New approaches are needed to overcome this hurdle and allow large scale expansion of human hepatocytes in immunodeficient swine.
Collapse
Affiliation(s)
- Erek David Nelson
- Mayo Clinic Minnesota, 4352, Surgery, 100 First St NW, Rochester, Rochester, Minnesota, United States, 55905-0002;
| | - Ellen Larson
- Mayo Clinic Minnesota, 4352, Surgery, Rochester, Minnesota, United States;
| | - Dong Jin Joo
- Mayo Clinic Minnesota, 4352, Surgery, Rochester, Minnesota, United States;
| | - Shennen Mao
- Mayo Clinic Minnesota, 4352, Surgery, Rochester, Minnesota, United States;
| | - Jaime Glorioso
- Mayo Clinic Minnesota, 4352, Surgery, Rochester, Minnesota, United States;
| | - Anan Abu Rmilah
- Mayo Clinic Minnesota, 4352, Surgery, Rochester, Minnesota, United States;
| | - Wei Zhou
- Mayo Clinic Minnesota, 4352, Surgery, Rochester, Minnesota, United States;
| | - Yao Jia
- Mayo Clinic Minnesota, 4352, Surgery, Rochester, Minnesota, United States;
| | - Taofic Mounajjed
- Mayo Clinic Minnesota, 4352, Laboratory Medicine and Pathology, Rochester, Minnesota, United States;
| | - Min Shi
- Mayo Clinic Minnesota, 4352, Laboratory Medicine and Pathology, Rochester, Minnesota, United States;
| | - Melanie Bois
- Mayo Clinic Minnesota, 4352, Laboratory Medicine and Pathology, Rochester, Minnesota, United States;
| | - Adam Wood
- Mayo Clinic Minnesota, 4352, Laboratory Medicine and Pathology, Rochester, Minnesota, United States;
| | - Fang Jin
- Mayo Clinic Minnesota, 4352, Immunology, Rochester, Minnesota, United States;
| | - Kristin Whitworth
- University of Missouri, 14716, National Swine Resource and Research Center, Division of Animal Sciences, Columbia, Missouri, United States;
| | - Kevin Wells
- University of Missouri, 14716, National Swine Resource and Research Center, Division of Animal Sciences, Columbia, Missouri, United States;
| | - Anna Spate
- University of Missouri, 14716, National Swine Resource and Research Center, Division of Animal Sciences, Columbia, Missouri, United States;
| | - Melissa Samuel
- University of Missouri, 14716, National Swine Resource and Research Center, Division of Animal Sciences, Columbia, Missouri, United States;
| | - Anna Minshew
- Mayo Clinic Minnesota, 4352, Surgery, Rochester, Minnesota, United States;
| | - Eric Walters
- University of Missouri, 14716, National Swine Resource and Research Center, Division of Animal Sciences, Columbia, Missouri, United States;
| | - Piero Rinaldo
- Mayo Clinic Minnesota, 4352, Laboratory Medicine and Pathology, Rochester, Minnesota, United States;
| | - Joeseph Lillegard
- Mayo Clinic Minnesota, 4352, Surgery, Rochester, Minnesota, United States;
| | - Aaron Johnson
- Mayo Clinic Minnesota, 4352, Immunology, Rochester, Minnesota, United States;
| | - Bruce Amiot
- Mayo Clinic Minnesota, 4352, Surgery, Rochester, Minnesota, United States;
| | - Raymond Hickey
- Mayo Clinic Minnesota, 4352, Surgery, Rochester, Minnesota, United States;
| | - Randall Prather
- University of Missouri, 14716, National Swine Resource and Research Center, Division of Animal Sciences, Columbia, Missouri, United States;
| | - Jeffrey L Platt
- University of Michigan Michigan Medicine, 21614, Surgery, Ann Arbor, Michigan, United States;
| | - Scott Lyle Nyberg
- Mayo Clinic Minnesota, 4352, Surgery, Rochester, Minnesota, United States;
| |
Collapse
|
22
|
Mullegama SV, Klein SD, Williams SR, Innis JW, Probst FJ, Haldeman-Englert C, Martinez-Agosto JA, Yang Y, Tian Y, Elsea SH, Ezashi T. Transcriptome analysis of MBD5-associated neurodevelopmental disorder (MAND) neural progenitor cells reveals dysregulation of autism-associated genes. Sci Rep 2021; 11:11295. [PMID: 34050248 PMCID: PMC8163803 DOI: 10.1038/s41598-021-90798-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 04/20/2021] [Indexed: 11/30/2022] Open
Abstract
MBD5-associated neurodevelopmental disorder (MAND) is an autism spectrum disorder (ASD) characterized by intellectual disability, motor delay, speech impairment and behavioral problems; however, the biological role of methyl-CpG-binding domain 5, MBD5, in neurodevelopment and ASD remains largely undefined. Hence, we created neural progenitor cells (NPC) derived from individuals with chromosome 2q23.1 deletion and conducted RNA-seq to identify differentially expressed genes (DEGs) and the biological processes and pathways altered in MAND. Primary skin fibroblasts from three unrelated individuals with MAND and four unrelated controls were converted into induced pluripotent stem cell (iPSC) lines, followed by directed differentiation of iPSC to NPC. Transcriptome analysis of MAND NPC revealed 468 DEGs (q < 0.05), including 20 ASD-associated genes. Comparison of DEGs in MAND with SFARI syndromic autism genes revealed a striking significant overlap in biological processes commonly altered in neurodevelopmental phenotypes, with TGFβ, Hippo signaling, DNA replication, and cell cycle among the top enriched pathways. Overall, these transcriptome deviations provide potential connections to the overlapping neurocognitive and neuropsychiatric phenotypes associated with key high-risk ASD genes, including chromatin modifiers and epigenetic modulators, that play significant roles in these disease states.
Collapse
Affiliation(s)
- Sureni V Mullegama
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
- Department of Molecular and Cellular Biology, College of Osteopathic Medicine, Sam Houston State University, Conroe, TX, 77304, USA
| | - Steven D Klein
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Medical Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | | | - Jeffrey W Innis
- Departments of Human Genetics, Pediatrics and Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Frank J Probst
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | | | - Julian A Martinez-Agosto
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ying Yang
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, 33612, USA
| | - Yuchen Tian
- Division of Animal Sciences and Bond Life Sciences Center, University of Missouri, Columbia, MO, 65211, USA
| | - Sarah H Elsea
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
| | - Toshihiko Ezashi
- Division of Animal Sciences and Bond Life Sciences Center, University of Missouri, Columbia, MO, 65211, USA.
| |
Collapse
|
23
|
Establishing an immunocompromised porcine model of human cancer for novel therapy development with pancreatic adenocarcinoma and irreversible electroporation. Sci Rep 2021; 11:7584. [PMID: 33828203 PMCID: PMC8027815 DOI: 10.1038/s41598-021-87228-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 03/25/2021] [Indexed: 02/07/2023] Open
Abstract
New therapies to treat pancreatic cancer are direly needed. However, efficacious interventions lack a strong preclinical model that can recapitulate patients’ anatomy and physiology. Likewise, the availability of human primary malignant tissue for ex vivo studies is limited. These are significant limitations in the biomedical device field. We have developed RAG2/IL2RG deficient pigs using CRISPR/Cas9 as a large animal model with the novel application of cancer xenograft studies of human pancreatic adenocarcinoma. In this proof-of-concept study, these pigs were successfully generated using on-demand genetic modifications in embryos, circumventing the need for breeding and husbandry. Human Panc01 cells injected subcutaneously into the ears of RAG2/IL2RG deficient pigs demonstrated 100% engraftment with growth rates similar to those typically observed in mouse models. Histopathology revealed no immune cell infiltration and tumor morphology was highly consistent with the mouse models. The electrical properties and response to irreversible electroporation of the tumor tissue were found to be similar to excised human pancreatic cancer tumors. The ample tumor tissue produced enabled improved accuracy and modeling of the electrical properties of tumor tissue. Together, this suggests that this model will be useful and capable of bridging the gap of translating therapies from the bench to clinical application.
Collapse
|
24
|
Humanization of Immunodeficient Animals for the Modeling of Transplantation, Graft Versus Host Disease, and Regenerative Medicine. Transplantation 2021; 104:2290-2306. [PMID: 32068660 PMCID: PMC7590965 DOI: 10.1097/tp.0000000000003177] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The humanization of animals is a powerful tool for the exploration of human disease pathogenesis in biomedical research, as well as for the development of therapeutic interventions with enhanced translational potential. Humanized models enable us to overcome biologic differences that exist between humans and other species, while giving us a platform to study human processes in vivo. To become humanized, an immune-deficient recipient is engrafted with cells, tissues, or organoids. The mouse is the most well studied of these hosts, with a variety of immunodeficient strains available for various specific uses. More recently, efforts have turned to the humanization of other animal species such as the rat, which offers some technical and immunologic advantages over mice. These advances, together with ongoing developments in the incorporation of human transgenes and additional mutations in humanized mouse models, have expanded our opportunities to replicate aspects of human allotransplantation and to assist in the development of immunotherapies. In this review, the immune and tissue humanization of various species is presented with an emphasis on their potential for use as models for allotransplantation, graft versus host disease, and regenerative medicine.
Collapse
|
25
|
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: 0.7] [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
|
26
|
Thomas AL, Taylor JS, Dunn JCY. Human skin-derived precursor cells xenografted in aganglionic bowel. J Pediatr Surg 2020; 55:2791-2796. [PMID: 32253016 DOI: 10.1016/j.jpedsurg.2020.03.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/21/2020] [Accepted: 03/14/2020] [Indexed: 12/23/2022]
Abstract
PURPOSE One in 5000 newborns is diagnosed with Hirschsprung disease each year in the United States. The potential of employing neural crest stem cells to restore the enteric nervous system has been investigated. Skin-derived precursor cells (SKPs) are multipotent progenitor cells that can differentiate into neurons and gliocytes in vitro and generate enteric ganglion-like structures in rodents. Here we examined the behavior of human SKPs (hSKPs) after their transplantation into a large animal model of colonic aganglionosis. METHODS Juvenile minipigs underwent a chemical denervation of the colon to establish an aganglionosis model. The hSKPs were generated from human foreskin and were cultured in neuroglial-selective medium. Cells were labeled with a fluorescent dye and were injected into the porcine aganglionic colon. After one week, transplanted hSKPs were assessed by immunofluorescence for markers of multipotency and neuroglial differentiation. RESULTS In culture, hSKPs expressed nestin and S100b indicative of neuroglial precursors. After xenografting in pigs, hSKPs were identified in the myenteric and submucosal plexuses of the colons. The hSKPs expressed nestin and early neuroglial differentiation markers. CONCLUSIONS Human SKPs transplanted into aganglionic colon demonstrated immunophenotypes of neuroglial progenitors, suggesting their potential use for Hirschsprung disease.
Collapse
Affiliation(s)
- Anne-Laure Thomas
- Division of Pediatric Surgery, Department of Surgery, School of Medicine, Stanford University, Division of Pediatric Surgery, 300 Pasteur Drive, Alway M116, Stanford, CA 94305
| | - Jordan S Taylor
- Division of Pediatric Surgery, Department of Surgery, School of Medicine, Stanford University, Division of Pediatric Surgery, 300 Pasteur Drive, Alway M116, Stanford, CA 94305
| | - James C Y Dunn
- Division of Pediatric Surgery, Department of Surgery, School of Medicine, Stanford University, Division of Pediatric Surgery, 300 Pasteur Drive, Alway M116, Stanford, CA 94305.
| |
Collapse
|
27
|
Lee K, Farrell K, Uh K. Application of genome-editing systems to enhance available pig resources for agriculture and biomedicine. Reprod Fertil Dev 2020; 32:40-49. [PMID: 32188556 DOI: 10.1071/rd19273] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Traditionally, genetic engineering in the pig was a challenging task. Genetic engineering of somatic cells followed by somatic cell nuclear transfer (SCNT) could produce genetically engineered (GE) pigs carrying site-specific modifications. However, due to difficulties in engineering the genome of somatic cells and developmental defects associated with SCNT, a limited number of GE pig models were reported. Recent developments in genome-editing tools, such as zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs) and the clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated (Cas) 9 system, have markedly changed the effort and time required to produce GE pig models. The frequency of genetic engineering in somatic cells is now practical. In addition, SCNT is no longer essential in producing GE pigs carrying site-specific modifications, because direct injection of genome-editing systems into developing embryos introduces targeted modifications. To date, the CRISPR/Cas9 system is the most convenient, cost-effective, timely and commonly used genome-editing technology. Several applicable biomedical and agricultural pig models have been generated using the CRISPR/Cas9 system. Although the efficiency of genetic engineering has been markedly enhanced with the use of genome-editing systems, improvements are still needed to optimally use the emerging technology. Current and future advances in genome-editing strategies will have a monumental effect on pig models used in agriculture and biomedicine.
Collapse
Affiliation(s)
- Kiho Lee
- Department of Animal and Poultry Sciences, Litton-Reaves Hall, Virginia Tech, Blacksburg, Virgina 24061, USA; and Corresponding author.
| | - Kayla Farrell
- Department of Animal and Poultry Sciences, Litton-Reaves Hall, Virginia Tech, Blacksburg, Virgina 24061, USA
| | - Kyungjun Uh
- Department of Animal and Poultry Sciences, Litton-Reaves Hall, Virginia Tech, Blacksburg, Virgina 24061, USA
| |
Collapse
|
28
|
Xenogeneic cross-circulation for extracorporeal recovery of injured human lungs. Nat Med 2020; 26:1102-1113. [PMID: 32661401 PMCID: PMC9990469 DOI: 10.1038/s41591-020-0971-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 06/09/2020] [Indexed: 12/15/2022]
Abstract
Patients awaiting lung transplantation face high wait-list mortality, as injury precludes the use of most donor lungs. Although ex vivo lung perfusion (EVLP) is able to recover marginal quality donor lungs, extension of normothermic support beyond 6 h has been challenging. Here we demonstrate that acutely injured human lungs declined for transplantation, including a lung that failed to recover on EVLP, can be recovered by cross-circulation of whole blood between explanted human lungs and a Yorkshire swine. This xenogeneic platform provided explanted human lungs a supportive, physiologic milieu and systemic regulation that resulted in functional and histological recovery after 24 h of normothermic support. Our findings suggest that cross-circulation can serve as a complementary approach to clinical EVLP to recover injured donor lungs that could not otherwise be utilized for transplantation, as well as a translational research platform for immunomodulation and advanced organ bioengineering.
Collapse
|
29
|
Iqbal MA, Hong K, Kim JH, Choi Y. Severe combined immunodeficiency pig as an emerging animal model for human diseases and regenerative medicines. BMB Rep 2020. [PMID: 31722780 PMCID: PMC6889892 DOI: 10.5483/bmbrep.2019.52.11.267] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Severe combined immunodeficiency (SCID) is a group of inherited disorders characterized by compromised T lymphocyte differentiation related to abnormal development of other lymphocytes [i.e., B and/or natural killer (NK) cells], leading to death early in life unless treated immediately with hematopoietic stem cell transplant. Functional NK cells may impact engraftment success of life-saving procedures such as bone marrow transplantation in human SCID patients. Therefore, in animal models, a T cell−/B cell−/NK cell+ environment provides a valuable tool for understanding the function of the innate immune system and for developing targeted NK therapies against human immune diseases. In this review, we focus on underlying mechanisms of human SCID, recent progress in the development of SCID animal models, and utilization of SCID pig model in biomedical sciences. Numerous physiologies in pig are comparable to those in human such as immune system, X-linked heritability, typical T−B+NK− cellular phenotype, and anatomy. Due to analogous features of pig to those of human, studies have found that immunodeficient pig is the most appropriate model for human SCID.
Collapse
Affiliation(s)
- Muhammad Arsalan Iqbal
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Center (SRC), Konkuk University, Seoul 05029, Korea
| | - Kwonho Hong
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Center (SRC), Konkuk University, Seoul 05029, Korea
| | - Jin Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Center (SRC), Konkuk University, Seoul 05029, Korea
| | - Youngsok Choi
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Center (SRC), Konkuk University, Seoul 05029, Korea
| |
Collapse
|
30
|
Boettcher AN, Li Y, Ahrens AP, Kiupel M, Byrne KA, Loving CL, Cino-Ozuna AG, Wiarda JE, Adur M, Schultz B, Swanson JJ, Snella EM, Ho CS(S, Charley SE, Kiefer ZE, Cunnick JE, Putz EJ, Dell'Anna G, Jens J, Sathe S, Goldman F, Westin ER, Dekkers JCM, Ross JW, Tuggle CK. Novel Engraftment and T Cell Differentiation of Human Hematopoietic Cells in ART-/-IL2RG-/Y SCID Pigs. Front Immunol 2020; 11:100. [PMID: 32117254 PMCID: PMC7017803 DOI: 10.3389/fimmu.2020.00100] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 01/15/2020] [Indexed: 01/08/2023] Open
Abstract
Pigs with severe combined immunodeficiency (SCID) are an emerging biomedical animal model. Swine are anatomically and physiologically more similar to humans than mice, making them an invaluable tool for preclinical regenerative medicine and cancer research. One essential step in further developing this model is the immunological humanization of SCID pigs. In this work we have generated T- B- NK- SCID pigs through site directed CRISPR/Cas9 mutagenesis of IL2RG within a naturally occurring DCLRE1C (ARTEMIS)-/- genetic background. We confirmed ART-/-IL2RG-/Y pigs lacked T, B, and NK cells in both peripheral blood and lymphoid tissues. Additionally, we successfully performed a bone marrow transplant on one ART-/-IL2RG-/Y male SCID pig with bone marrow from a complete swine leukocyte antigen (SLA) matched donor without conditioning to reconstitute porcine T and NK cells. Next, we performed in utero injections of cultured human CD34+ selected cord blood cells into the fetal ART-/-IL2RG-/Y SCID pigs. At birth, human CD45+ CD3ε+ cells were detected in cord and peripheral blood of in utero injected SCID piglets. Human leukocytes were also detected within the bone marrow, spleen, liver, thymus, and mesenteric lymph nodes of these animals. Taken together, we describe critical steps forwards the development of an immunologically humanized SCID pig model.
Collapse
Affiliation(s)
| | - Yunsheng Li
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Amanda P. Ahrens
- Laboratory Animal Resources, 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
| | - Kristen A. Byrne
- Food Safety and Enteric Pathogen Unit, National Animal Disease Center, US Department of Agriculture, Agricultural Research Service, Ames, IA, United States
| | - Crystal L. Loving
- Food Safety and Enteric Pathogen Unit, National Animal Disease Center, US Department of Agriculture, Agricultural Research Service, Ames, IA, United States
| | - A. Giselle Cino-Ozuna
- Veterinary Diagnostic Laboratory, Kansas State University, Manhattan, KS, United States
| | - Jayne E. Wiarda
- Food Safety and Enteric Pathogen Unit, National Animal Disease Center, US Department of Agriculture, Agricultural Research Service, Ames, IA, United States
- Immunobiology Graduate Program, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
- Oak Ridge Institute for Science and Education, Agricultural Research Service Participation Program, Oak Ridge, TN, United States
| | - Malavika Adur
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Blythe Schultz
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | | | - Elizabeth M. Snella
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Chak-Sum (Sam) Ho
- Gift of Hope Organ and Tissue Donor Network, Itasca, IL, United States
| | - Sara E. Charley
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Zoe E. Kiefer
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Joan E. Cunnick
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Ellie J. Putz
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Giuseppe Dell'Anna
- Laboratory Animal Resources, Iowa State University, Ames, IA, United States
| | - Jackie Jens
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Swanand Sathe
- Veterinary Clinical Sciences, Iowa State University, Ames, IA, United States
| | - Frederick Goldman
- Department of Pediatrics, University of Alabama, Birmingham, AL, United States
| | - Erik R. Westin
- Department of Pediatrics, University of Alabama, Birmingham, AL, United States
| | - Jack C. M. Dekkers
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Jason W. Ross
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | | |
Collapse
|
31
|
Lee K, Uh K, Farrell K. Current progress of genome editing in livestock. Theriogenology 2020; 150:229-235. [PMID: 32000993 DOI: 10.1016/j.theriogenology.2020.01.036] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 01/18/2020] [Indexed: 12/12/2022]
Abstract
Historically, genetic engineering in livestock proved to be challenging. Without stable embryonic stem cell lines to utilize, somatic cell nuclear transfer (SCNT) had to be employed to produce many of the genetically engineered (GE) livestock models. Through the genetic engineering of somatic cells followed by SCNT, GE livestock models could be generated carrying site-specific modifications. Although successful, only a few GE livestock models were generated because of low efficiency and associated birth defects. Recently, there have been major strides in the development of genome editing tools: Zinc-Finger Nucleases (ZFNs), Transcription activator-like effector nucleases (TALENS), and Clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated 9 (Cas9) system. These tools rely on the generation of a double strand DNA break, followed by one of two repair pathways: non-homologous end joining (NHEJ) or homology directed repair (HDR). Compared to the traditional approaches, these tools dramatically reduce time and effort needed to establish a GE animal. Another benefit of utilizing genome editing tools is the application of direct injection into developing embryos to induce targeted mutations, therefore, eliminating side effects associated with SCNT. Emerging technological advancements of genome editing systems have dramatically improved efficiency to generate GE livestock models for both biomedical and agricultural purposes. Although the efficiency of genome editing tools has revolutionized GE livestock production, improvements for safe and consistent application are desired. This review will provide an overview of genome editing techniques, as well as examples of GE livestock models for agricultural and biomedical purposes.
Collapse
Affiliation(s)
- Kiho Lee
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, USA.
| | - Kyungjun Uh
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, USA
| | - Kayla Farrell
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, USA
| |
Collapse
|
32
|
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: 6.3] [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
|
33
|
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: 6.0] [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
|
34
|
Platt JL, Cascalho M, Piedrahita JA. Xenotransplantation: Progress Along Paths Uncertain from Models to Application. ILAR J 2019; 59:286-308. [PMID: 30541147 DOI: 10.1093/ilar/ily015] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 08/23/2018] [Indexed: 12/18/2022] Open
Abstract
For more than a century, transplantation of tissues and organs from animals into man, xenotransplantation, has been viewed as a potential way to treat disease. Ironically, interest in xenotransplantation was fueled especially by successful application of allotransplantation, that is, transplantation of human tissue and organs, as a treatment for a variety of diseases, especially organ failure because scarcity of human tissues limited allotransplantation to a fraction of those who could benefit. In principle, use of animals such as pigs as a source of transplants would allow transplantation to exert a vastly greater impact than allotransplantation on medicine and public health. However, biological barriers to xenotransplantation, including immunity of the recipient, incompatibility of biological systems, and transmission of novel infectious agents, are believed to exceed the barriers to allotransplantation and presently to hinder clinical applications. One way potentially to address the barriers to xenotransplantation is by genetic engineering animal sources. The last 2 decades have brought progressive advances in approaches that can be applied to genetic modification of large animals. Application of these approaches to genetic engineering of pigs has contributed to dramatic improvement in the outcome of experimental xenografts in nonhuman primates and have encouraged the development of a new type of xenograft, a reverse xenograft, in which human stem cells are introduced into pigs under conditions that support differentiation and expansion into functional tissues and potentially organs. These advances make it appropriate to consider the potential limitation of genetic engineering and of current models for advancing the clinical applications of xenotransplantation and reverse xenotransplantation.
Collapse
Affiliation(s)
- Jeffrey L Platt
- Surgery, Microbiology & Immunology, and Transplantation Biology, University of Michigan, Ann Arbor, Michigan
| | - Marilia Cascalho
- Surgery, Microbiology & Immunology, and Transplantation Biology, University of Michigan, Ann Arbor, Michigan
| | - Jorge A Piedrahita
- Translational Medicine and The Comparative Medicine Institute, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| |
Collapse
|
35
|
Putz EJ, Putz AM, Boettcher A, Charley S, Sauer M, Palmer M, Phillips R, Hostetter J, Loving CL, Cunnick JE, Tuggle CK. Successful development of methodology for detection of hapten-specific contact hypersensitivity (CHS) memory in swine. PLoS One 2019; 14:e0223483. [PMID: 31596901 PMCID: PMC6785115 DOI: 10.1371/journal.pone.0223483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 09/23/2019] [Indexed: 11/30/2022] Open
Abstract
Hapten contact hypersensitivity (CHS) elicits a well-documented inflammation response that can be used to illustrate training of immune cells through hapten-specific CHS memory. The education of hapten-specific memory T cells has been well-established, recent research in mice has expanded the “adaptive” characteristic of a memory response from solely a function of the adaptive immune system, to innate cells as well. To test whether similar responses are seen in a non-rodent model, we used hapten-specific CHS to measure the ear inflammation response of outbred pigs to dinitrofluorobenzene (DNFB), oxazolone (OXA), or vehicle controls. We adapted mouse innate memory literature protocols to the domestic pig model. Animals were challenged up to 32 days post initial sensitization exposure to the hapten, and specific ear swelling responses to this challenge were significant for 7, 21, and 32 days post-sensitization. We established hapten-specific CHS memory exists in a non-rodent model. We also developed a successful protocol for demonstrating these CHS responses in a porcine system.
Collapse
Affiliation(s)
- E. J. Putz
- Iowa State University, Department of Animal Science, Ames, IA, United States of America
| | - A. M. Putz
- Iowa State University, Department of Animal Science, Ames, IA, United States of America
| | - A. Boettcher
- Iowa State University, Department of Animal Science, Ames, IA, United States of America
| | - S. Charley
- Iowa State University, Department of Animal Science, Ames, IA, United States of America
| | - M. Sauer
- Iowa State University, Department of Animal Science, Ames, IA, United States of America
| | - M. Palmer
- USDA-ARS-National Animal Disease Center, Infectious Bacterial Diseases of Livestock Research Unit, Ames, IA, United States of America
| | - R. Phillips
- Iowa State University College of Veterinary Medicine, Department of Veterinary Pathology Science, Ames, IA, United States of America
| | - J. Hostetter
- Iowa State University College of Veterinary Medicine, Department of Veterinary Pathology Science, Ames, IA, United States of America
| | - C. L. Loving
- USDA-ARS-National Animal Disease Center, Food Safety and Enteric Pathogens Research Unit, Ames, IA, United States of America
| | - J. E. Cunnick
- Iowa State University, Department of Animal Science, Ames, IA, United States of America
| | - C. K. Tuggle
- Iowa State University, Department of Animal Science, Ames, IA, United States of America
- * E-mail:
| |
Collapse
|
36
|
|
37
|
Abstract
We present the most recent research results on the creation of pigs that can accept human cells. Pigs in which grafted human cells can flourish are essential for studies of the production of human organs in the pig and for verification of the efficacy of cells and tissues of human origin for use in regenerative therapy. First, against the background of a worldwide shortage of donor organs, the need for future medical technology to produce human organs for transplantation is discussed. We then describe proof-of-concept studies in small animals used to produce human organs. An overview of the history of studies examining the induction of immune tolerance by techniques involving fertilized animal eggs and the injection of human cells into fetuses or neonatal animals is also presented. Finally, current and future prospects for producing pigs that can accept human cells and tissues for experimental purposes are discussed.
Collapse
|
38
|
Yuan Y, Park J, Tian Y, Choi J, Pasquariello R, Alexenko AP, Dai A, Behura SK, Roberts RM, Ezashi T. A six-inhibitor culture medium for improving naïve-type pluripotency of porcine pluripotent stem cells. Cell Death Discov 2019; 5:104. [PMID: 31240131 PMCID: PMC6579764 DOI: 10.1038/s41420-019-0184-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 05/28/2019] [Indexed: 02/06/2023] Open
Abstract
Understanding essential signaling network requirements and making appropriate adjustments in culture conditions are crucial if porcine pluripotent stem cells (PSC) are to achieve their full potential. Here, we first used two protein factors (LIF and FGF2) and kinase inhibitor combinations in attempts to convert primed type lentiviral-reprogrammed porcine induced PSC (Lv-piPSC) into naïve-like state and developed a medium called FL6i. In addition to FGF2 and LIF, this medium contained inhibitors of MAPK14, MAPK8, TGFB1, MAP2K1, GSK3A and BMP. Crucially, the usual TGFB1 and BMP4 protein components of many stem cell media were replaced in FL6i with inhibitors of TGFB1 and BMP. With this medium, Lv-piPSC were readily transformed from their original primed state into cells that formed colonies with typical features of naïve-state stem cells. The FL6i medium also assisted generation of naïve-type piPSC lines from porcine embryonic fibroblasts with non-integrating episomal plasmids (Epi-piPSC). These lines, despite retaining variable amounts of vector DNA, expressed higher endogenous pPOU5F1 and pSOX2 than Lv-piPSC. They have been cultured without obvious morphological change for >45 passages and retained pluripotent phenotypes in terms of upregulation of genes associated with pluripotency, low expression of genes linked to emergence of somatic cell lineages, and ability to generate well differentiated teratomas in immune-compromised mice. FL6i conditions, therefore, appear to support elevated pluripotent phenotypes. However, FL6i was less able to support the generation of embryonic stem cells from porcine blastocysts. Although colonies with dome-shaped morphologies were evident and the cells had some gene expression features linked to pluripotency, the phenotypes were ultimately not stable. Pathway analysis derived from RNAseq data performed on the various cell lines generated in this study suggest the benefits of employing the FL6i medium on porcine cells reside in its ability to minimize TGFB1 and BMP signaling, which would otherwise de-stabilize the stem cell state.
Collapse
Affiliation(s)
- Ye Yuan
- 1Bond Life Sciences Center, University of Missouri, Columbia, MO 65211 USA.,2Division of Animal Sciences, University of Missouri, Columbia, MO 65211 USA.,3Colorado Center for Reproductive Medicine, Lone Tree, CO 80124 USA
| | - Jinkyu Park
- 1Bond Life Sciences Center, University of Missouri, Columbia, MO 65211 USA.,2Division of Animal Sciences, University of Missouri, Columbia, MO 65211 USA.,4Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510 USA
| | - Yuchen Tian
- 1Bond Life Sciences Center, University of Missouri, Columbia, MO 65211 USA
| | - Jungmin Choi
- 5Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY 10065 USA
| | - Rolando Pasquariello
- 3Colorado Center for Reproductive Medicine, Lone Tree, CO 80124 USA.,6Department of Agricultural and Environmental Sciences-Production, Landscape, Agroenergy, University of Milan, Milano, 20133 Italy
| | - Andrei P Alexenko
- 1Bond Life Sciences Center, University of Missouri, Columbia, MO 65211 USA.,2Division of Animal Sciences, University of Missouri, Columbia, MO 65211 USA
| | - Aihua Dai
- 1Bond Life Sciences Center, University of Missouri, Columbia, MO 65211 USA
| | - Susanta K Behura
- 2Division of Animal Sciences, University of Missouri, Columbia, MO 65211 USA
| | - R Michael Roberts
- 1Bond Life Sciences Center, University of Missouri, Columbia, MO 65211 USA.,2Division of Animal Sciences, University of Missouri, Columbia, MO 65211 USA
| | - Toshihiko Ezashi
- 1Bond Life Sciences Center, University of Missouri, Columbia, MO 65211 USA.,2Division of Animal Sciences, University of Missouri, Columbia, MO 65211 USA
| |
Collapse
|
39
|
Carey K, Ryu J, Uh K, Lengi AJ, Clark-Deener S, Corl BA, Lee K. Frequency of off-targeting in genome edited pigs produced via direct injection of the CRISPR/Cas9 system into developing embryos. BMC Biotechnol 2019; 19:25. [PMID: 31060546 PMCID: PMC6501304 DOI: 10.1186/s12896-019-0517-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 04/23/2019] [Indexed: 12/24/2022] Open
Abstract
Background The CRISPR/Cas9 system can effectively introduce site-specific modifications to the genome. The efficiency is high enough to induce targeted genome modifications during embryogenesis, thus increasing the efficiency of producing genetically modified animal models and having potential clinical applications as an assisted reproductive technology. Because most of the CRISPR/Cas9 systems introduce site-specific double-stranded breaks (DSBs) to induce site-specific modifications, a major concern is its potential off-targeting activity, which may hinder the application of the technology in clinics. In this study, we investigated off-targeting events in genome edited pigs/fetuses that were generated through direct injection of the CRISPR/Cas9 system into developing embryos; off-targeting activity of four different sgRNAs targeting RAG2, IL2RG, SCD5, and Ig Heavy chain were examined. Results First, bioinformatics analysis was applied to identify 27 potential off-targeting genes from the sgRNAs. Then, PCR amplification followed by sequencing analysis was used to verify the presence of off-targeting events. Off-targeting events were only identified from the sgRNA used to disrupt Ig Heavy chain in pigs; frequency of off-targeting was 80 and 70% on AR and RBFOX1 locus respectively. A potential PAM sequence was present in both of the off-targeting genes adjacent to probable sgRNA binding sites. Mismatches against sgRNA were present only on the 5′ side of AR, suggesting that off-targeting activities are systematic events. However, the mismatches on RBFOX1 were not limited to the 5′ side, indicating unpredictability of the events. Conclusions The prevalence of off-targeting is low via direct injection of CRISPR/Cas9 system into developing embryos, but the events cannot be accurately predicted. Off-targeting frequency of each CRISPR/Cas9 system should be deliberately assessed prior to its application in clinics. Electronic supplementary material The online version of this article (10.1186/s12896-019-0517-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kayla Carey
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Junghyun Ryu
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Kyungjun Uh
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Andrea J Lengi
- Department of Dairy Science, Virginia Tech, Blacksburg, VA, 24061, USA
| | | | - Benjamin A Corl
- Department of Dairy Science, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Kiho Lee
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, 24061, USA.
| |
Collapse
|
40
|
Boettcher AN, Ahrens AP, Charley SE, Tuggle CK. A Comprehensive Protocol for Laparotomy in Swine to Facilitate Ultrasound-Guided Injection into the Fetal Intraperitoneal Space. Comp Med 2019; 69:123-129. [PMID: 30755290 DOI: 10.30802/aalas-cm-18-000098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Swine are a commonly used animal model for biomedical research. One research application of swine models is the in utero injection of human or pig cells into the fetal liver (FL) or intraperitoneal space. In utero injections can be accomplished through laparotomy procedures in pregnant swine. In this study, we aimed to establish comprehensive laparotomy protocols for ultrasound-guided injections into fetuses. Two pregnant gilts, with a total of 16 fetuses, underwent laparotomy at 41 and 42 d of gestation. During surgery, we attempted to inject half of the fetuses in the FL or intraperitoneal space with saline and titanium wire for radiographic imaging after birth. After the laparotomy and fetal injections, both gilts maintained pregnancy throughout gestation and initiated labor at full term. Of the 16 fetuses present at the time of laparotomy, 12 were liveborn, 2 were stillborn, and the remaining 2 were mummies. A total of 7 fetuses from the 2 litters were known to have been injected with a wire during the surgery. After farrowing, piglets were radiographed, and 6 piglets were identified to have wire within the abdominal space. Livers were dissected, and additional radiographs were obtained. It was determined that one piglet had wire within the liver, whereas the other 5 had wire within the intraperitoneal space. Overall, we describe in-depth laparotomy surgery protocols, ultrasound-guided injection of saline and titanium wire into the FL or intraperitoneal space, postoperative monitoring protocols, and information on radiographic detection of titanium wire after piglet birth. These protocols can be followed by other research groups intending to inject cells of interest into either the intraperitoneal space or FL of fetal piglets.
Collapse
Affiliation(s)
| | - Amanda P Ahrens
- Laboratory Animal Resources, Iowa State University, Ames, Iowa
| | - Sara E Charley
- Department of Animal Science, Iowa State University, Ames, Iowa
| | | |
Collapse
|
41
|
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: 3.0] [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
|
42
|
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: 4.3] [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
|
43
|
Boettcher AN, Cunnick JE, Powell EJ, Egner TK, Charley SE, Loving CL, Tuggle CK. Porcine signal regulatory protein alpha binds to human CD47 to inhibit phagocytosis: Implications for human hematopoietic stem cell transplantation into severe combined immunodeficient pigs. Xenotransplantation 2018; 26:e12466. [PMID: 30311702 DOI: 10.1111/xen.12466] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 08/10/2018] [Accepted: 09/18/2018] [Indexed: 12/27/2022]
Abstract
BACKGROUND Severe combined immunodeficient (SCID) pigs are an emerging animal model being developed for biomedical and regenerative medicine research. SCID pigs can successfully engraft human-induced pluripotent stem cells and cancer cell lines. The development of a humanized SCID pig through xenotransplantation of human hematopoietic stem cells (HSCs) would be a further demonstration of the value of such a large animal SCID model. Xenotransplantation success with HSCs into non-obese diabetic (NOD)-derived SCID mice is dependent on the ability of NOD mouse signal regulatory protein alpha (SIRPA) to bind human CD47, inducing higher phagocytic tolerance than other mouse strains. Therefore, we investigated whether porcine SIRPA binds human CD47 in the context of developing a humanized SCID pig. METHODS Peripheral blood mononuclear cells (PBMCs) were collected from SCID and non-SCID pigs. Flow cytometry was used to assess whether porcine monocytes could bind to human CD47. Porcine monocytes were isolated from PBMCs and were subjected to phagocytosis assays with pig, human, and mouse red blood cell (RBC) targets. Blocking phagocytosis assays were performed by incubating human RBCs with anti-human CD47 blocking antibody B6H12, non-blocking antibody 2D3, and nonspecific IgG1 antibody and exposing to human or porcine monocytes. RESULTS We found that porcine SIRPA binds to human CD47 in vitro by flow cytometric assays. Additionally, phagocytosis assays were performed, and we found that porcine monocytes phagocytose human and porcine RBCs at significantly lower levels than mouse RBCs. When human RBCs were preincubated with CD47 antibodies B6H12 or 2D3, phagocytosis was induced only after B6H12 incubation, indicating the lower phagocytic activity of porcine monocytes with human cells requires interaction between porcine SIRPA and human CD47. CONCLUSIONS We have shown the first evidence that porcine monocytes can bind to human CD47 and are phagocytically tolerant to human cells, suggesting that porcine SCID models have the potential to support engraftment of human HSCs.
Collapse
Affiliation(s)
| | - Joan E Cunnick
- Department of Animal Science, Iowa State University, Ames, Iowa
| | - Ellis J Powell
- Department of Animal Science, Iowa State University, Ames, Iowa.,National Animal Disease Center, Ruminant Diseases and Immunology Unit, US Department of Agriculture, Agricultural Research Service, Ames, Iowa
| | | | - Sara E Charley
- Department of Animal Science, Iowa State University, Ames, Iowa
| | - Crystal L Loving
- National Animal Disease Center, Food Safety and Enteric Pathogens Unit, US Department of Agriculture, Agricultural Research Service, Ames, Iowa
| | | |
Collapse
|
44
|
Yang H, Wu Z. Genome Editing of Pigs for Agriculture and Biomedicine. Front Genet 2018; 9:360. [PMID: 30233645 PMCID: PMC6131568 DOI: 10.3389/fgene.2018.00360] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 08/21/2018] [Indexed: 12/26/2022] Open
Abstract
Pigs serve as an important agricultural resource and animal model in biomedical studies. Efficient and precise modification of pig genome by using recently developed gene editing tools has significantly broadened the application of pig models in various research areas. The three types of site-specific nucleases, namely, zinc-finger nucleases, transcription activator-like effector nucleases, and clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein, are the main gene editing tools that can efficiently introduce predetermined modifications, including knockouts and knockins, into the pig genome. These modifications can confer desired phenotypes to pigs to improve production traits, such as optimal meat production, enhanced feed digestibility, and disease resistance. Besides, given their genetic, anatomic, and physiologic similarities to humans, pigs can also be modified to model human diseases or to serve as an organ source for xenotransplantation to save human lives. To date, many genetically modified pig models with agricultural or biomedical values have been established by using gene editing tools. These pig models are expected to accelerate research progress in related fields and benefit humans.
Collapse
Affiliation(s)
- Huaqiang Yang
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Zhenfang Wu
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
45
|
Powell EJ, Charley S, Boettcher A, Varley L, Brown J, Schroyen M, Adur MK, Dekkers S, Isaacson D, Sauer M, Cunnick J, Ellinwood NM, Ross JW, Dekkers J, Tuggle C. Creating effective biocontainment facilities and maintenance protocols for raising specific pathogen-free, severe combined immunodeficient (SCID) pigs. Lab Anim 2018; 52:402-412. [PMID: 29325489 PMCID: PMC7737622 DOI: 10.1177/0023677217750691] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Severe combined immunodeficiency (SCID) is defined by the lack of an adaptive immune system. Mutations causing SCID are found naturally in humans, mice, horses, dogs, and recently in pigs, with the serendipitous discovery of the Iowa State University SCID pigs. As research models, SCID animals are naturally tolerant of xenotransplantation and offer valuable insight into research areas such as regenerative medicine, cancer therapy, as well as immune cell signaling mechanisms. Large-animal biomedical models, particularly pigs, are increasingly essential to advance the efficacy and safety of novel regenerative therapies on human disease. Thus, there is a need to create practical approaches to maintain hygienic severe immunocompromised porcine models for exploratory medical research. Such research often requires stable genetic lines for replication and survival of healthy SCID animals for months post-treatment. A further hurdle in the development of the ISU SCID pig as a biomedical model involved the establishment of facilities and protocols necessary to obtain clean SPF piglets from the conventional pig farm on which they were discovered. A colony of homozygous SCID boars and SPF carrier sows has been created and maintained through selective breeding, bone marrow transplants, innovative husbandry techniques, and the development of biocontainment facilities.
Collapse
Affiliation(s)
| | - Sara Charley
- Department of Animal Science, Iowa State University
| | | | - Lisa Varley
- Department of Animal Science, Iowa State University
| | | | | | | | | | | | - Mary Sauer
- Laboratory Animal Resources, Iowa State University
| | - Joan Cunnick
- Department of Animal Science, Iowa State University
| | | | | | - Jack Dekkers
- Department of Animal Science, Iowa State University
| | | |
Collapse
|
46
|
Cho B, Kim SJ, Lee EJ, Ahn SM, Lee JS, Ji DY, Lee K, Kang JT. Generation of insulin-deficient piglets by disrupting INS gene using CRISPR/Cas9 system. Transgenic Res 2018; 27:289-300. [PMID: 29691708 DOI: 10.1007/s11248-018-0074-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 04/17/2018] [Indexed: 12/13/2022]
Abstract
Diabetes mellitus is a chronic disease with accompanying severe complications. Various animal models, mostly rodents due to availability of genetically modified lines, have been used to investigate the pathophysiology of diabetes. Using pigs for diabetic research can be beneficial because of their similarity in size, pathogenesis pathway, physiology, and metabolism with human. However, the use of pigs for diabetes research has been hampered due to only few pig models presenting diabetes symptoms. In this study, we have successfully generated insulin-deficient pigs by generating the indels of the porcine INS gene in somatic cells using CRISPR/Cas9 system followed by somatic cell nuclear transfer. First, somatic cells carrying a modified INS gene were generated using CRISPR/Cas9 system and their genotypes were confirmed by T7E1 assay; targeting efficiency was 40.4% (21/52). After embryo transfer, three live and five stillborn piglets were born. As expected, INS knockout piglets presented high blood glucose levels and glucose was detected in the urine. The level of insulin and c-peptide in the blood serum of INS knockout piglets were constant after feeding and the expression of insulin in the pancreas was absent in those piglets. This study demonstrates effectiveness of CRISPR/Cas9 system in generating novel pig models. We expect that these insulin-deficient pigs can be used in diabetes research to test the efficacy and safety of new drugs and the recipient of islet transplantation to investigate optimal transplantation strategies.
Collapse
Affiliation(s)
- Bumrae Cho
- Biotechnology Research Institute, Mgenplus Co., Ltd., Seoul, Korea
| | - Su Jin Kim
- Biotechnology Research Institute, Mgenplus Co., Ltd., Seoul, Korea
| | - Eun-Jin Lee
- Biotechnology Research Institute, Mgenplus Co., Ltd., Seoul, Korea
| | - Sun Mi Ahn
- Biotechnology Research Institute, Mgenplus Co., Ltd., Seoul, Korea
| | - Jin Seok Lee
- Biotechnology Research Institute, Mgenplus Co., Ltd., Seoul, Korea
| | - Dal-Young Ji
- Biotechnology Research Institute, Mgenplus Co., Ltd., Seoul, Korea
| | - Kiho Lee
- Department of Animal and Poultry Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA
| | - Jung-Taek Kang
- Biotechnology Research Institute, Mgenplus Co., Ltd., Seoul, Korea.
| |
Collapse
|
47
|
Use of gene-editing technology to introduce targeted modifications in pigs. J Anim Sci Biotechnol 2018; 9:5. [PMID: 29423214 PMCID: PMC5787920 DOI: 10.1186/s40104-017-0228-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 12/22/2017] [Indexed: 01/06/2023] Open
Abstract
Pigs are an important resource in agriculture and serve as a model for human diseases. Due to their physiological and anatomical similarities with humans, pigs can recapitulate symptoms of human diseases, making them a useful model in biomedicine. However, in the past pig models have not been widely used partially because of the difficulty in genetic modification. The lack of true embryonic stem cells in pigs forced researchers to utilize genetic modification in somatic cells and somatic cell nuclear transfer (SCNT) to generate genetically engineered (GE) pigs carrying site-specific modifications. Although possible, this approach is extremely inefficient and GE pigs born through this method often presented developmental defects associated with the cloning process. Advancement in the gene-editing systems such as Zinc-Finger Nucleases (ZFNs), Transcription activator-like effector nucleases (TALENs), and the Clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated 9 (Cas9) system have dramatically increased the efficiency of producing GE pigs. These gene-editing systems, specifically engineered endonucleases, are based on inducing double-stranded breaks (DSBs) at a specific location, and then site-specific modifications can be introduced through one of the two DNA repair pathways: non-homologous end joining (NHEJ) or homology direct repair (HDR). Random insertions or deletions (indels) can be introduced through NHEJ and specific nucleotide sequences can be introduced through HDR, if donor DNA is provided. Use of these engineered endonucleases provides a higher success in genetic modifications, multiallelic modification of the genome, and an opportunity to introduce site-specific modifications during embryogenesis, thus bypassing the need of SCNT in GE pig production. This review will provide a historical prospective of GE pig production and examples of how the gene-editing system, led by engineered endonucleases, have improved GE pig production. We will also present some of our current progress related to the optimal use of CRISPR/Cas9 system during embryogenesis.
Collapse
|
48
|
Shen Y, Xu K, Yuan Z, Guo J, Zhao H, Zhang X, Zhao L, Qing Y, Li H, Pan W, Jia B, Zhao HY, Wei HJ. Efficient generation of P53 biallelic knockout Diannan miniature pigs via TALENs and somatic cell nuclear transfer. J Transl Med 2017; 15:224. [PMID: 29100547 PMCID: PMC5670695 DOI: 10.1186/s12967-017-1327-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 10/27/2017] [Indexed: 11/13/2022] Open
Abstract
Background Pigs have many features that make them attractive as biomedical models for various diseases, including cancer. P53 is an important tumor suppressor gene that exerts a central role in protecting cells from oncogenic transformation and is mutated in a large number of human cancers. P53 mutations occur in almost every type of tumor and in over 50% of all tumors. In a recent publication, pigs with a mutated P53 gene were generated that resulted in lymphoma and renal and osteogenic tumors. However, approximately 80% of human tumors have dysfunctional P53. A P53-deficient pig model is still required to elucidate. Methods Transcription activator-like effector nucleases (TALENs) were designed to target porcine P53 exon 4. The targeting activity was evaluated using a luciferase SSA recombination assay. P53 biallelic knockout (KO) cell lines were established from single-cell colonies of fetal fibroblasts derived from Diannan miniature pigs followed by electroporation with TALENs plasmids. One cell line was selected as the donor cell line for somatic cell nuclear transfer (SCNT) for the generation of P53 KO pigs. P53 KO stillborn fetuses and living piglets were obtained. Gene typing of the collected cloned individuals was performed by T7EI assay and sequencing. Fibroblast cells from Diannan miniature piglets with a P53 biallelic knockout or wild type were analyzed for the P53 response to doxorubicin treatment by confocal microscopy and western blotting. Results The luciferase SSA recombination assay revealed that the targeting activities of the designed TALENs were 55.35-fold higher than those of the control. Eight cell lines (8/19) were mutated for P53, and five of them were biallelic knockouts. One of the biallelic knockout cell lines was selected as nuclear donor cells for SCNT. The cloned embryos were transferred into five recipient gilts, three of them becoming pregnant. Five live fetuses were obtained from one surrogate by caesarean section after 38 days of gestation for genotyping. Finally, six live piglets and one stillborn piglet were collected from two recipients by caesarean section. Sequencing analyses of the target site confirmed the P53 biallelic knockout in all fetuses and piglets, consistent with the genotype of the donor cells. The qPCR analysis showed that the expression of the P53 mRNA had significant reduction in various tissues of the knockout piglets. Furthermore, confocal microscopy and western blotting analyses demonstrated that the fibroblast cells of Diannan miniature piglets with a P53 biallelic knockout were defective in mediating DNA damage when incubated with doxorubicin. Conclusion TALENs combined with SCNT was successfully used to generate P53 KO Diannan miniature pigs. Although these genetically engineered Diannan miniature pigs had no tumorigenic signs, the P53 gene was dysfunctional. We believe that these pigs will provide powerful new resources for preclinical oncology and basic cancer research. Electronic supplementary material The online version of this article (10.1186/s12967-017-1327-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Youfeng Shen
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China.,College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Kaixiang Xu
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China
| | - Zaimei Yuan
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China.,College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Jianxiong Guo
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China
| | - Heng Zhao
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China.,College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Xuezeng Zhang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China.,Key Laboratory Animal Nutrition and Feed of Yunnan Province, Yunnan Agricultural University, Kunming, 650201, China
| | - Lu Zhao
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China.,Key Laboratory Animal Nutrition and Feed of Yunnan Province, Yunnan Agricultural University, Kunming, 650201, China
| | - Yubo Qing
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China.,College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Honghui Li
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China.,College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Weirong Pan
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Baoyu Jia
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China.,College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Hong-Ye Zhao
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China.
| | - Hong-Jiang Wei
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China. .,College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China. .,Key Laboratory Animal Nutrition and Feed of Yunnan Province, Yunnan Agricultural University, Kunming, 650201, China.
| |
Collapse
|
49
|
Genome editing in livestock: Are we ready for a revolution in animal breeding industry? Transgenic Res 2017; 26:715-726. [DOI: 10.1007/s11248-017-0049-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Accepted: 10/24/2017] [Indexed: 12/25/2022]
|
50
|
Stem cells and genome editing: approaches to tissue regeneration and regenerative medicine. J Hum Genet 2017; 63:165-178. [PMID: 29192237 DOI: 10.1038/s10038-017-0348-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/20/2017] [Accepted: 08/21/2017] [Indexed: 12/20/2022]
Abstract
Understanding the basis of regeneration of each tissue and organ, and incorporating this knowledge into clinical treatments for degenerative tissues and organs in patients, are major goals for researchers in regenerative biology. Here we provide an overview of current work, from high-regeneration animal models, to stem cell-based culture models, transplantation technologies, large-animal chimeric models, and programmable nuclease-based genome-editing technologies. Three-dimensional culture generating organoids, which represents intact tissue/organ identity including cell fate and morphology are getting more general approaches in the fields by taking advantage of embryonic stem cells, induced pluripotent stem cells and adult stem cells. The organoid culture system potentially has profound impact on the field of regenerative medicine. We also emphasize that the large animal model, in particular pig model would be a hope to manufacture humanized organs in in vivo empty (vacant) niche, which now potentially allows not only appropriate cell fate identity but nearly the same property as human organs in size. Therefore, integrative and collaborative researches across different fields might be critical to the aims needed in clinical trial.
Collapse
|