1
|
Han Y, Zhou J, Zhang R, Liang Y, Lai L, Li Z. Genome-edited rabbits: Unleashing the potential of a promising experimental animal model across diverse diseases. Zool Res 2024; 45:253-262. [PMID: 38287906 PMCID: PMC11017087 DOI: 10.24272/j.issn.2095-8137.2023.201] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 12/05/2023] [Indexed: 01/31/2024] Open
Abstract
Animal models are extensively used in all aspects of biomedical research, with substantial contributions to our understanding of diseases, the development of pharmaceuticals, and the exploration of gene functions. The field of genome modification in rabbits has progressed slowly. However, recent advancements, particularly in CRISPR/Cas9-related technologies, have catalyzed the successful development of various genome-edited rabbit models to mimic diverse diseases, including cardiovascular disorders, immunodeficiencies, aging-related ailments, neurological diseases, and ophthalmic pathologies. These models hold great promise in advancing biomedical research due to their closer physiological and biochemical resemblance to humans compared to mice. This review aims to summarize the novel gene-editing approaches currently available for rabbits and present the applications and prospects of such models in biomedicine, underscoring their impact and future potential in translational medicine.
Collapse
Affiliation(s)
- Yang Han
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Science, Jilin University, Changchun, Jilin 130062, China
| | - Jiale Zhou
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Science, Jilin University, Changchun, Jilin 130062, China
| | - Renquan Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Science, Jilin University, Changchun, Jilin 130062, China
| | - Yuru Liang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Science, Jilin University, Changchun, Jilin 130062, China
| | - Liangxue Lai
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Science, Jilin University, Changchun, Jilin 130062, China
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530, China
- Guangzhou Regenerative Medicine and Health Guang Dong Laboratory (GRMH-GDL), Guangzhou, Guangdong 510005, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China. E-mail:
| | - Zhanjun Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal Science, Jilin University, Changchun, Jilin 130062, China. E-mail:
| |
Collapse
|
2
|
Lei K, Zhang W, Chen J, McKinney SA, Ross EJ, Lee HC, Sánchez Alvarado A. Pluripotency retention and exogenous mRNA introduction in planarian stem cells in culture. iScience 2023; 26:106001. [PMID: 36866042 PMCID: PMC9971864 DOI: 10.1016/j.isci.2023.106001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/31/2022] [Accepted: 01/13/2023] [Indexed: 01/21/2023] Open
Abstract
Planarians possess naturally occurring pluripotent adult somatic stem cells (neoblasts) required for homeostasis and whole-body regeneration. However, no reliable neoblast culture methods are currently available, hindering mechanistic studies of pluripotency and the development of transgenic tools. We report robust methods for neoblast culture and delivery of exogenous mRNAs. We identify optimal culture media for the short-term maintenance of neoblasts in vitro and show via transplantation that cultured stem cells retain pluripotency for two days. We developed a procedure that significantly improves neoblast yield and purity by modifying standard flow cytometry methods. These methods enable the introduction and expression of exogenous mRNAs in neoblasts, overcoming a key hurdle impeding the application of transgenics in planarians. The advances in cell culture reported here create new opportunities for mechanistic studies of planarian adult stem cell pluripotency, and provide a systematic framework to develop cell culture techniques in other emerging research organisms.
Collapse
Affiliation(s)
- Kai Lei
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
- Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China
| | - Wenya Zhang
- Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China
| | - Jiajia Chen
- Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang 310024, China
| | - Sean A. McKinney
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Eric J. Ross
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
- Howard Hughes Medical Institute, Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Heng-Chi Lee
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
| | - Alejandro Sánchez Alvarado
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
- Howard Hughes Medical Institute, Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| |
Collapse
|
3
|
Yokoyama T, Imai K, Hashimoto Y. Comparison of cell viability between mouse-derived ES-D3 cells and Balb/c 3T3 cells using denture-base lining materials. Dent Mater J 2022; 41:481-486. [PMID: 35264546 DOI: 10.4012/dmj.2021-302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The study was done to compare cell viability between ES-D3 and Balb/c 3T3 cells, and evaluate the difference in cell viability between these cell lines using denture-base lining materials for prosthetic dentistry. To compare the cytotoxicity, three acrylic and three silicone dental materials were used. The cell viability was examined by MTT and lactate dehydrogenase (LDH) methods. The cell viability immediately after malaxation or light irradiation was lower only for the acrylic materials in 3T3 cells, and for both silicone and acrylic materials in ES-D3 cells. However, the cell viability determined 24 h after malaxation or light irradiation by the MTT and LDH methods did not significantly differ between samples. It was observed that ES-D3 cells are more sensitive depending on the type of material. The results suggest that ES-D3 cells can be used as in vitro systems for conducting biosafety assessment to predict embryotoxicity.
Collapse
Affiliation(s)
- Tadashi Yokoyama
- Department of Biomaterials, School of Dentistry, Osaka Dental University
| | - Koichi Imai
- Department of Tissue Engineering, School of Dentistry, Osaka Dental University.,School of Health Sciences, Osaka Dental University
| | - Yoshiya Hashimoto
- Department of Biomaterials, School of Dentistry, Osaka Dental University
| |
Collapse
|
4
|
Soto DA, Navarro M, Zheng C, Halstead MM, Zhou C, Guiltinan C, Wu J, Ross PJ. Simplification of culture conditions and feeder-free expansion of bovine embryonic stem cells. Sci Rep 2021; 11:11045. [PMID: 34040070 PMCID: PMC8155104 DOI: 10.1038/s41598-021-90422-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 05/05/2021] [Indexed: 02/04/2023] Open
Abstract
Bovine embryonic stem cells (bESCs) extend the lifespan of the transient pluripotent bovine inner cell mass in vitro. After years of research, derivation of stable bESCs was only recently reported. Although successful, bESC culture relies on complex culture conditions that require a custom-made base medium and mouse embryonic fibroblasts (MEF) feeders, limiting the widespread use of bESCs. We report here simplified bESC culture conditions based on replacing custom base medium with a commercially available alternative and eliminating the need for MEF feeders by using a chemically-defined substrate. bESC lines were cultured and derived using a base medium consisting of N2B27 supplements and 1% BSA (NBFR-bESCs). Newly derived bESC lines were easy to establish, simple to propagate and stable after long-term culture. These cells expressed pluripotency markers and actively proliferated for more than 35 passages while maintaining normal karyotype and the ability to differentiate into derivatives of all three germ lineages in embryoid bodies and teratomas. In addition, NBFR-bESCs grew for multiple passages in a feeder-free culture system based on vitronectin and Activin A medium supplementation while maintaining pluripotency. Simplified conditions will facilitate the use of bESCs for gene editing applications and pluripotency and lineage commitment studies.
Collapse
Affiliation(s)
- Delia Alba Soto
- Department of Animal Science, University of California, 450 Bioletti Way, Davis, CA, 95616, USA
| | - Micaela Navarro
- Department of Animal Science, University of California, 450 Bioletti Way, Davis, CA, 95616, USA
- Instituto de Investigaciones Biotecnológicas 'Dr Rodolfo Ugalde', UNSAM-CONICET, Buenos Aires, Argentina
| | - Canbin Zheng
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390, USA
| | | | - Chuan Zhou
- Department of Animal Science, University of California, 450 Bioletti Way, Davis, CA, 95616, USA
| | - Carly Guiltinan
- Department of Animal Science, University of California, 450 Bioletti Way, Davis, CA, 95616, USA
| | - Jun Wu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390, USA
| | - Pablo Juan Ross
- Department of Animal Science, University of California, 450 Bioletti Way, Davis, CA, 95616, USA.
| |
Collapse
|
5
|
Xu J, Zhang J, Yang D, Song J, Pallas B, Zhang C, Hu J, Peng X, Christensen ND, Han R, Chen YE. Gene Editing in Rabbits: Unique Opportunities for Translational Biomedical Research. Front Genet 2021; 12:642444. [PMID: 33584832 PMCID: PMC7876448 DOI: 10.3389/fgene.2021.642444] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 01/04/2021] [Indexed: 11/13/2022] Open
Abstract
The rabbit is a classic animal model for biomedical research, but the production of gene targeted transgenic rabbits had been extremely challenging until the recent advent of gene editing tools. More than fifty gene knockout or knock-in rabbit models have been reported in the past decade. Gene edited (GE) rabbit models, compared to their counterpart mouse models, may offer unique opportunities in translational biomedical research attributed primarily to their relatively large size and long lifespan. More importantly, GE rabbit models have been found to mimic several disease pathologies better than their mouse counterparts particularly in fields focused on genetically inherited diseases, cardiovascular diseases, ocular diseases, and others. In this review we present selected examples of research areas where GE rabbit models are expected to make immediate contributions to the understanding of the pathophysiology of human disease, and support the development of novel therapeutics.
Collapse
Affiliation(s)
- Jie Xu
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Jifeng Zhang
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Dongshan Yang
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Jun Song
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Brooke Pallas
- Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Chen Zhang
- Biomedical Sciences and Biophysics Graduate Program, Division of Cardiac Surgery, Department of Surgery, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Jiafen Hu
- Department of Pathology and Laboratory Medicine, Penn State Cancer Institute, Hershey, PA, United States
| | - Xuwen Peng
- Department of Comparative Medicine, Penn State University College of Medicine, Hershey, PA, United States
| | - Neil D Christensen
- Department of Pathology and Laboratory Medicine, Penn State Cancer Institute, Hershey, PA, United States.,Department of Microbiology and Immunology, Penn State University College of Medicine, Hershey, PA, United States
| | - Renzhi Han
- Biomedical Sciences and Biophysics Graduate Program, Division of Cardiac Surgery, Department of Surgery, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Y Eugene Chen
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
6
|
Song J, Zhang J, Xu J, Garcia-Barrio M, Chen YE, Yang D. Genome engineering technologies in rabbits. J Biomed Res 2021; 35:135-147. [PMID: 32934190 PMCID: PMC8038526 DOI: 10.7555/jbr.34.20190133] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The rabbit has been recognized as a valuable model in various biomedical and biological research fields because of its intermediate size and phylogenetic proximity to primates. However, the technology for precise genome manipulations in rabbit has been stalled for decades, severely limiting its applications in biomedical research. Novel genome editing technologies, especially CRISPR/Cas9, have remarkably enhanced precise genome manipulation in rabbits, and shown their superiority and promise for generating rabbit models of human genetic diseases. In this review, we summarize the brief history of transgenic rabbit technology and the development of novel genome editing technologies in rabbits.
Collapse
Affiliation(s)
- Jun Song
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Jifeng Zhang
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Jie Xu
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Minerva Garcia-Barrio
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Y Eugene Chen
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Dongshan Yang
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| |
Collapse
|
7
|
Heintz N, Gong S. Working with Bacterial Artificial Chromosomes (BACs) and Other High-Capacity Vectors. Cold Spring Harb Protoc 2020; 2020:2020/10/pdb.top097998. [PMID: 33004554 DOI: 10.1101/pdb.top097998] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Genetic targeting of specific cell types is fundamentally important for modern molecular-genetic studies. The development of simple methods to engineer high-capacity vectors-in particular, bacterial artificial chromosomes (BACs)-for the preparation of transgenic lines that accurately express a gene of interest has resulted in commonplace usage of transgenic techniques in a wide variety of experimental systems. Here we provide a brief description of each of the four major types of large-capacity vectors, with a focus on the use of BAC vectors.
Collapse
|
8
|
Abstract
The mouse is one of the most widely used model organisms for genetic study. The tools available to alter the mouse genome have developed over the preceding decades from forward screens to gene targeting in stem cells to the recent influx of CRISPR approaches. In this review, we first consider the history of mice in genetic study, the development of classic approaches to genome modification, and how such approaches have been used and improved in recent years. We then turn to the recent surge of nuclease-mediated techniques and how they are changing the field of mouse genetics. Finally, we survey common classes of alleles used in mice and discuss how they might be engineered using different methods.
Collapse
Affiliation(s)
- James F Clark
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mt. Sinai, New York, New York 10029, USA
| | - Colin J Dinsmore
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mt. Sinai, New York, New York 10029, USA
| | - Philippe Soriano
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mt. Sinai, New York, New York 10029, USA
| |
Collapse
|
9
|
Imai H, Tsuda S, Iwamori T, Kano K, Kusakabe KT, Ono E. Establishment of a novel method for the production of chimeric mouse embryos using water-in-oil droplets. Exp Anim 2020; 70:84-90. [PMID: 32999214 PMCID: PMC7887616 DOI: 10.1538/expanim.20-0060] [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] [Indexed: 11/04/2022] Open
Abstract
Production of chimeric animals is often a necessity for the generation of genetically
modified animals and has gained popularity in recent years in regenerative medicine for
the reconstruction of xenogeneic organs. Aggregation and injection methods are generally
used to produce chimeric mice. In the aggregation method, the chimeras are produced by
co-culturing embryos and stem cells, and keeping them physically adhered, although it may
not be an assured method for producing chimeric embryos. In the injection method, the
chimeras are produced by injecting stem cells into the zona pellucida using
microcapillaries; however, this technique requires a high degree of skill. This study
aimed to establish a novel method for producing chimeric embryos via water-in-oil droplets
that differs from conventional methods. In this study, embryonic stem cells and embryos
were successfully isolated in the droplets, and the emergence of chimeric embryos was
confirmed by co-culture for 6 h. Using this method, the control and operability of stem
cell numbers could be regulated, and reproducibility and quantification were improved
during the production of chimeric embryos. In addition to the conventional methods for
producing chimeric embryos, the novel method described here could be employed for the
efficient production of chimeric animals.
Collapse
Affiliation(s)
- Hiroyuki Imai
- Department of Biomedicine, Graduate School of Medical Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.,Laboratory of Veterinary Anatomy, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi 753-8515, Japan
| | - Soichiro Tsuda
- On-chip Biotechnologies Co., Ltd., 2-24-16 Naka-cho, Koganei, Tokyo 184-0012, Japan
| | - Tokuko Iwamori
- Department of Biomedicine, Graduate School of Medical Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.,Laboratory of Zoology, Graduate School of Agriculture, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Kiyoshi Kano
- Laboratory of Veterinary Developmental Biology, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi 753-8515, Japan
| | - Ken Takeshi Kusakabe
- Laboratory of Veterinary Anatomy, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi 753-8515, Japan
| | - Etsuro Ono
- Department of Biomedicine, Graduate School of Medical Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
10
|
Seita Y, Tsukiyama T, Azami T, Kobayashi K, Iwatani C, Tsuchiya H, Nakaya M, Tanabe H, Hitoshi S, Miyoshi H, Nakamura S, Kawauchi A, Ema M. Comprehensive evaluation of ubiquitous promoters suitable for the generation of transgenic cynomolgus monkeys†. Biol Reprod 2020; 100:1440-1452. [PMID: 30869744 DOI: 10.1093/biolre/ioz040] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 02/21/2019] [Accepted: 03/12/2019] [Indexed: 12/11/2022] Open
Abstract
Nonhuman primates (NHPs) are considered to be the most valuable models for human transgenic (Tg) research into disease because human pathology is more closely recapitulated in NHPs than rodents. Previous studies have reported the generation of Tg NHPs that ubiquitously overexpress a transgene using various promoters, but it is not yet clear which promoter is most suitable for the generation of NHPs overexpressing a transgene ubiquitously and persistently in various tissues. To clarify this issue, we evaluated four putative ubiquitous promoters, cytomegalovirus (CMV) immediate-early enhancer and chicken beta-actin (CAG), elongation factor 1α (EF1α), ubiquitin C (UbC), and CMV, using an in vitro differentiation system of cynomolgus monkey embryonic stem cells (ESCs). While the EF1α promoter drove Tg expression more strongly than the other promoters in undifferentiated pluripotent ESCs, the CAG promoter was more effective in differentiated cells such as embryoid bodies and ESC-derived neurons. When the CAG and EF1α promoters were used to generate green fluorescent protein (GFP)-expressing Tg monkeys, the CAG promoter drove GFP expression in skin and hematopoietic tissues more strongly than in ΕF1α-GFP Tg monkeys. Notably, the EF1α promoter underwent more silencing in both ESCs and Tg monkeys. Thus, the CAG promoter appears to be the most suitable for ubiquitous and stable expression of transgenes in the differentiated tissues of Tg cynomolgus monkeys and appropriate for the establishment of human disease models.
Collapse
Affiliation(s)
- Yasunari Seita
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Tomoyuki Tsukiyama
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Shiga, Japan.,Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Takuya Azami
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Kenichi Kobayashi
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Shiga, Japan.,Department of Urology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Chizuru Iwatani
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Hideaki Tsuchiya
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Masataka Nakaya
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Hideyuki Tanabe
- Department of Evolutionary Studies of Biosystems, School of Advanced Sciences, SOKENDAI (The Graduate University for Advanced Studies), Hayama, Kanagawa, Japan
| | - Seiji Hitoshi
- Department of Physiology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Hiroyuki Miyoshi
- Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Shinichiro Nakamura
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Akihiro Kawauchi
- Department of Urology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Masatsugu Ema
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Shiga, Japan.,Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Sakyo-ku, Kyoto, Japan.,PRESTO, Japan Science and Technology Agency, Honcho, Saitama, Japan
| |
Collapse
|
11
|
Fischer I, Dulin JN, Lane MA. Transplanting neural progenitor cells to restore connectivity after spinal cord injury. Nat Rev Neurosci 2020; 21:366-383. [PMID: 32518349 PMCID: PMC8384139 DOI: 10.1038/s41583-020-0314-2] [Citation(s) in RCA: 168] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2020] [Indexed: 12/12/2022]
Abstract
Spinal cord injury remains a scientific and therapeutic challenge with great cost to individuals and society. The goal of research in this field is to find a means of restoring lost function. Recently we have seen considerable progress in understanding the injury process and the capacity of CNS neurons to regenerate, as well as innovations in stem cell biology. This presents an opportunity to develop effective transplantation strategies to provide new neural cells to promote the formation of new neuronal networks and functional connectivity. Past and ongoing clinical studies have demonstrated the safety of cell therapy, and preclinical research has used models of spinal cord injury to better elucidate the underlying mechanisms through which donor cells interact with the host and thus increase long-term efficacy. While a variety of cell therapies have been explored, we focus here on the use of neural progenitor cells obtained or derived from different sources to promote connectivity in sensory, motor and autonomic systems.
Collapse
Affiliation(s)
- Itzhak Fischer
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA.
| | - Jennifer N Dulin
- Department of Biology, Texas A&M University, College Station, TX, USA
| | - Michael A Lane
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
12
|
García-García MJ. A History of Mouse Genetics: From Fancy Mice to Mutations in Every Gene. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1236:1-38. [PMID: 32304067 DOI: 10.1007/978-981-15-2389-2_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The laboratory mouse has become the model organism of choice in numerous areas of biological and biomedical research, including the study of congenital birth defects. The appeal of mice for these experimental studies stems from the similarities between the physiology, anatomy, and reproduction of these small mammals with our own, but it is also based on a number of practical reasons: mice are easy to maintain in a laboratory environment, are incredibly prolific, and have a relatively short reproductive cycle. Another compelling reason for choosing mice as research subjects is the number of tools and resources that have been developed after more than a century of working with these small rodents in laboratory environments. As will become obvious from the reading of the different chapters in this book, research in mice has already helped uncover many of the genes and processes responsible for congenital birth malformations and human diseases. In this chapter, we will provide an overview of the methods, scientific advances, and serendipitous circumstances that have made these discoveries possible, with a special emphasis on how the use of genetics has propelled scientific progress in mouse research and paved the way for future discoveries.
Collapse
|
13
|
Landel C, Pritchett-Corning KR. Gene Editing Technologies and Use of Recombinant/Synthetic Nucleic Acids in Laboratory Animals. APPLIED BIOSAFETY 2018. [DOI: 10.1177/1535676018797353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
14
|
Vieira MS, Santos AK, Vasconcellos R, Goulart VAM, Parreira RC, Kihara AH, Ulrich H, Resende RR. Neural stem cell differentiation into mature neurons: Mechanisms of regulation and biotechnological applications. Biotechnol Adv 2018; 36:1946-1970. [PMID: 30077716 DOI: 10.1016/j.biotechadv.2018.08.002] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 07/31/2018] [Accepted: 08/01/2018] [Indexed: 02/07/2023]
Abstract
The abilities of stem cells to self-renew and form different mature cells expand the possibilities of applications in cell-based therapies such as tissue recomposition in regenerative medicine, drug screening, and treatment of neurodegenerative diseases. In addition to stem cells found in the embryo, various adult organs and tissues have niches of stem cells in an undifferentiated state. In the central nervous system of adult mammals, neurogenesis occurs in two regions: the subventricular zone and the dentate gyrus in the hippocampus. The generation of the different neural lines originates in adult neural stem cells that can self-renew or differentiate into astrocytes, oligodendrocytes, or neurons in response to specific stimuli. The regulation of the fate of neural stem cells is a finely controlled process relying on a complex regulatory network that extends from the epigenetic to the translational level and involves extracellular matrix components. Thus, a better understanding of the mechanisms underlying how the process of neurogenesis is induced, regulated, and maintained will provide elues for development of novel for strategies for neurodegenerative therapies. In this review, we focus on describing the mechanisms underlying the regulation of the neuronal differentiation process by transcription factors, microRNAs, and extracellular matrix components.
Collapse
Affiliation(s)
- Mariana S Vieira
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Instituto Nanocell, Divinopólis, MG, Brazil
| | - Anderson K Santos
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rebecca Vasconcellos
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Instituto Nanocell, Divinopólis, MG, Brazil
| | - Vânia A M Goulart
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ricardo C Parreira
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Instituto Nanocell, Divinopólis, MG, Brazil
| | - Alexandre H Kihara
- Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, SP, Brazil.
| | - Rodrigo R Resende
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Instituto Nanocell, Divinopólis, MG, Brazil.
| |
Collapse
|
15
|
Esquerda-Canals G, Montoliu-Gaya L, Güell-Bosch J, Villegas S. Mouse Models of Alzheimer's Disease. J Alzheimers Dis 2018; 57:1171-1183. [PMID: 28304309 DOI: 10.3233/jad-170045] [Citation(s) in RCA: 191] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that nowadays affects more than 40 million people worldwide and it is predicted to exponentially increase in the coming decades. Because no curative treatment exists, research on the pathophysiology of the disease, as well as the testing of new drugs, are mandatory. For these purposes, animal models constitute a valuable, although perfectible tool. This review takes a tour through several aspects of mouse models of AD, such as the generation of transgenic models, the relevance of the promoter driving the expression of the transgenes, and the concrete transgenes used to simulate AD pathophysiology. Then, transgenic mouse lines harboring mutated human genes at several loci such as APP, PSEN1, APOEɛ4, and ob (leptin) are reviewed. Therefore, not only the accumulation of the Aβ peptide is emulated but also cholesterol and insulin metabolism. Further novel information about the disease will allow for the development of more accurate animal models, which in turn will undoubtedly be helpful for bringing preclinical research closer to clinical trials in humans.
Collapse
Affiliation(s)
- Gisela Esquerda-Canals
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.,Departament de Biologia Cel·lular, Fisiologia i Immunologia, Unitat de Citologia i Histologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Laia Montoliu-Gaya
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Jofre Güell-Bosch
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Sandra Villegas
- Protein Folding and Stability Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| |
Collapse
|
16
|
Klimczewska K, Kasperczuk A, Suwińska A. The Regulative Nature of Mammalian Embryos. Curr Top Dev Biol 2018; 128:105-149. [DOI: 10.1016/bs.ctdb.2017.10.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
17
|
Martin Gonzalez J, Morgani SM, Bone RA, Bonderup K, Abelchian S, Brakebusch C, Brickman JM. Embryonic Stem Cell Culture Conditions Support Distinct States Associated with Different Developmental Stages and Potency. Stem Cell Reports 2017; 7:177-91. [PMID: 27509134 PMCID: PMC4983099 DOI: 10.1016/j.stemcr.2016.07.009] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 07/08/2016] [Accepted: 07/10/2016] [Indexed: 11/29/2022] Open
Abstract
Embryonic stem cells (ESCs) are cell lines derived from the mammalian pre-implantation embryo. Here we assess the impact of derivation and culture conditions on both functional potency and ESC transcriptional identity. Individual ESCs cultured in either two small-molecule inhibitors (2i) or with knockout serum replacement (KOSR), but not serum, can generate high-level chimeras regardless of how these cells were derived. ESCs cultured in these conditions showed a transcriptional correlation with early pre-implantation embryos (E1.5–E3.5) and contributed to development from the 2-cell stage. Conversely, the transcriptome of serum-cultured ESCs correlated with later stages of development (E4.5), at which point embryonic cells are more restricted in their developmental potential. Thus, ESC culture systems are not equivalent, but support cell types that resemble distinct developmental stages. Cells derived in one condition can be reprogrammed to another developmental state merely by adaptation to another culture condition. ESC derivation condition does not irreversibly affect functional potency ESCs cultured in 2i and KOSR resemble early stages of embryonic development ESCs cultured in 2i and KOSR have enhanced functional potency ESCs cultured in KOSR resemble primitive endoderm
Collapse
Affiliation(s)
- Javier Martin Gonzalez
- Transgenic Core Facility, University of Copenhagen, 3B Blegdamsvej, 2200 Copenhagen N, Denmark
| | - Sophie M Morgani
- The Danish Stem Cell Centre - DanStem, University of Copenhagen, 3B Blegdamsvej, 2200 Copenhagen N, Denmark
| | - Robert A Bone
- The Danish Stem Cell Centre - DanStem, University of Copenhagen, 3B Blegdamsvej, 2200 Copenhagen N, Denmark
| | - Kasper Bonderup
- Transgenic Core Facility, University of Copenhagen, 3B Blegdamsvej, 2200 Copenhagen N, Denmark
| | - Sahar Abelchian
- Transgenic Core Facility, University of Copenhagen, 3B Blegdamsvej, 2200 Copenhagen N, Denmark
| | - Cord Brakebusch
- Transgenic Core Facility, University of Copenhagen, 3B Blegdamsvej, 2200 Copenhagen N, Denmark
| | - Joshua M Brickman
- The Danish Stem Cell Centre - DanStem, University of Copenhagen, 3B Blegdamsvej, 2200 Copenhagen N, Denmark.
| |
Collapse
|
18
|
Zhang Z, Zhang Y, Gao F, Han S, Cheah KS, Tse HF, Lian Q. CRISPR/Cas9 Genome-Editing System in Human Stem Cells: Current Status and Future Prospects. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 9:230-241. [PMID: 29246302 PMCID: PMC5651489 DOI: 10.1016/j.omtn.2017.09.009] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Revised: 09/27/2017] [Accepted: 09/27/2017] [Indexed: 12/21/2022]
Abstract
Genome-editing involves the insertion, deletion, or replacement of DNA in the genome of a living organism using “molecular scissors.” Traditional genome editing with engineered nucleases for human stem cells is limited by its low efficiency, high cost, and poor specificity. The CRISPR system has recently emerged as a powerful gene manipulation technique with advantages of high editing efficiency and low cost. Although this technique offers huge potential for gene manipulation in various organisms ranging from prokaryotes to higher mammals, there remain many challenges in human stem cell research. In this review, we highlight the basic biology and application of the CRISPR/Cas9 system in current human stem cell research, discuss its advantages and challenges, and debate the future prospects for human stem cells in regenerative medicine.
Collapse
Affiliation(s)
- Zhao Zhang
- Department of Medicine, University of Hong Kong, Hong Kong, China
| | - Yuelin Zhang
- Department of Medicine, University of Hong Kong, Hong Kong, China
| | - Fei Gao
- Department of Medicine, University of Hong Kong, Hong Kong, China
| | - Shuo Han
- Department of Medicine, University of Hong Kong, Hong Kong, China
| | - Kathryn S Cheah
- School of Biomedical Sciences, University of Hong Kong, Hong Kong, China
| | - Hung-Fat Tse
- Department of Medicine, University of Hong Kong, Hong Kong, China; Shenzhen Institutes of Research and Innovation, University of Hong Kong, Shenzhen, China
| | - Qizhou Lian
- Department of Medicine, University of Hong Kong, Hong Kong, China; Shenzhen Institutes of Research and Innovation, University of Hong Kong, Shenzhen, China; School of Biomedical Sciences, University of Hong Kong, Hong Kong, China.
| |
Collapse
|
19
|
Bai DP, Yang MM, Qu L, Chen YL. Generation of a transgenic cashmere goat using the piggyBac transposition system. Theriogenology 2017; 93:1-6. [DOI: 10.1016/j.theriogenology.2017.01.036] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Revised: 01/19/2017] [Accepted: 01/21/2017] [Indexed: 01/04/2023]
|
20
|
Bevacqua RJ, Fernandez-Martin R, Canel NG, Gibbons A, Texeira D, Lange F, Vans Landschoot G, Savy V, Briski O, Hiriart MI, Grueso E, Ivics Z, Taboga O, Kues WA, Ferraris S, Salamone DF. Assessing Tn5 and Sleeping Beauty for transpositional transgenesis by cytoplasmic injection into bovine and ovine zygotes. PLoS One 2017; 12:e0174025. [PMID: 28301581 PMCID: PMC5354444 DOI: 10.1371/journal.pone.0174025] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 01/06/2017] [Indexed: 12/27/2022] Open
Abstract
Transgenic domestic animals represent an alternative to bioreactors for large-scale production of biopharmaceuticals and could also provide more accurate biomedical models than rodents. However, their generation remains inefficient. Recently, DNA transposons allowed improved transgenesis efficiencies in mice and pigs. In this work, Tn5 and Sleeping Beauty (SB) transposon systems were evaluated for transgenesis by simple cytoplasmic injection in livestock zygotes. In the case of Tn5, the transposome complex of transposon nucleic acid and Tn5 protein was injected. In the case of SB, the supercoiled plasmids encoding a transposon and the SB transposase were co-injected. In vitro produced bovine zygotes were used to establish the cytoplasmic injection conditions. The in vitro cultured blastocysts were evaluated for reporter gene expression and genotyped. Subsequently, both transposon systems were injected in seasonally available ovine zygotes, employing transposons carrying the recombinant human factor IX driven by the beta-lactoglobulin promoter. The Tn5 approach did not result in transgenic lambs. In contrast, the Sleeping Beauty injection resulted in 2 lambs (29%) carrying the transgene. Both animals exhibited cellular mosaicism of the transgene. The extraembryonic tissues (placenta or umbilical cord) of three additional animals were also transgenic. These results show that transpositional transgenesis by cytoplasmic injection of SB transposon components can be applied for the production of transgenic lambs of pharmaceutical interest.
Collapse
Affiliation(s)
- R. J. Bevacqua
- Animal Biotechnology Laboratory, Facultad de Agronomia. INPA-CONICET, Buenos Aires University, Buenos Aires, Argentina
| | - R. Fernandez-Martin
- Animal Biotechnology Laboratory, Facultad de Agronomia. INPA-CONICET, Buenos Aires University, Buenos Aires, Argentina
| | - N. G. Canel
- Animal Biotechnology Laboratory, Facultad de Agronomia. INPA-CONICET, Buenos Aires University, Buenos Aires, Argentina
| | - A. Gibbons
- Experimental Station Bariloche, INTA, Bariloche, Argentina
| | - D. Texeira
- Laboratorio de Fisiologia e Controle da Reprodução, FAVET, UECE, Ceará State, Brasil
| | - F. Lange
- Cloning and Transgenesis Laboratory, Maimónides University, Buenos Aires, Argentina
| | - G. Vans Landschoot
- Animal Biotechnology Laboratory, Facultad de Agronomia. INPA-CONICET, Buenos Aires University, Buenos Aires, Argentina
- Cloning and Transgenesis Laboratory, Maimónides University, Buenos Aires, Argentina
| | - V. Savy
- Animal Biotechnology Laboratory, Facultad de Agronomia. INPA-CONICET, Buenos Aires University, Buenos Aires, Argentina
| | - O. Briski
- Animal Biotechnology Laboratory, Facultad de Agronomia. INPA-CONICET, Buenos Aires University, Buenos Aires, Argentina
| | - M. I. Hiriart
- Animal Biotechnology Laboratory, Facultad de Agronomia. INPA-CONICET, Buenos Aires University, Buenos Aires, Argentina
| | - E. Grueso
- Paul-Ehrlich-Institute, Langen, Germany
| | - Z. Ivics
- Paul-Ehrlich-Institute, Langen, Germany
| | - O. Taboga
- CICVyA Biotechnology Institute, INTA Castelar, Buenos Aires, Argentina
| | - W. A. Kues
- Friedrich-Loeffler-Institut, Neustadt, Germany
| | - S. Ferraris
- Cloning and Transgenesis Laboratory, Maimónides University, Buenos Aires, Argentina
| | - D. F. Salamone
- Animal Biotechnology Laboratory, Facultad de Agronomia. INPA-CONICET, Buenos Aires University, Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
21
|
Abstract
In the past few years, new technologies have arisen that enable higher efficiency of gene editing. With the increase ease of using gene editing technologies, it is important to consider the best method for transferring new genetic material to livestock animals. Microinjection is a technique that has proven to be effective in mice but is less efficient in large livestock animals. Over the years, a variety of methods have been used for cloning as well as gene transfer including; nuclear transfer, sperm mediated gene transfer (SMGT), and liposome-mediated DNA transfer. This review looks at the different success rate of these methods and how they have evolved to become more efficient. As well as gene editing technologies, including Zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and the most recent clustered regulatory interspaced short palindromic repeats (CRISPRs). Through the advancements in gene-editing technologies, generating transgenic animals is now more accessible and affordable. The goals of producing transgenic animals are to 1) increase our understanding of biology and biomedical science; 2) increase our ability to produce more efficient animals; and 3) produce disease resistant animals. ZFNs, TALENs, and CRISPRs combined with gene transfer methods increase the possibility of achieving these goals.
Collapse
Key Words
- BLG, β-lactoglobulin
- CRISPR
- CRISPRs, clustered regulatory interspaced short palindromic repeats
- EG, embryonic germ
- ES, Embryonic stem
- ESC, Embryonic stem cell
- HDR, homology directed repair
- ICM, inner cell mass
- ICSI, intracytoplasmic sperm injection
- NHEJ, non-homologous end joining
- NT, nuclear transfers
- OBCT, oocyte bisection technique
- PAM, protospacer adjacent motif
- PCR, polymerase chain reaction
- PGCS, primordial germ cells
- RVDs, repeat variable diresidues
- SMGT
- SMGT, sperm mediated gene transfer
- SV40, simian virus 40
- TALEN
- TALENs, transcription activator-like effector nucleases
- ZFN
- ZFN, Zinc-finger nucleases
- gene editing
- gene transfer
- iPSC, induced pluripotent stem cells
- nuclear transfer
- ssODN, single strand oligo nucleotide
Collapse
Affiliation(s)
- Samantha N Lotti
- a Department of Animal Sciences , University of Illinois at Urbana-Champaign , Urbana , Illinois , USA
| | - Kathryn M Polkoff
- a Department of Animal Sciences , University of Illinois at Urbana-Champaign , Urbana , Illinois , USA
| | - Marcello Rubessa
- b Carl R. Woese Institute for Genomic Biology, University of Illinois , Urbana , IL , USA
| | - Matthew B Wheeler
- a Department of Animal Sciences , University of Illinois at Urbana-Champaign , Urbana , Illinois , USA.,b Carl R. Woese Institute for Genomic Biology, University of Illinois , Urbana , IL , USA
| |
Collapse
|
22
|
Demain AL, Vandamme EJ, Collins J, Buchholz K. History of Industrial Biotechnology. Ind Biotechnol (New Rochelle N Y) 2016. [DOI: 10.1002/9783527807796.ch1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Arnold L. Demain
- Drew University; Charles A. Dana Research Institute for Scientists Emeriti (R.I.S.E.); 36, Madison Ave Madison NJ 07940 USA
| | - Erick J. Vandamme
- Ghent University; Department of Biochemical and Microbial Technology; Belgium
| | - John Collins
- Science historian; Leipziger Straße 82A; 38124 Braunschweig Germany
| | - Klaus Buchholz
- Technical University Braunschweig; Institute of Chemical Engineering; Hans-Sommer-Str. 10 38106 Braunschweig Germany
| |
Collapse
|
23
|
Schilit SLP, Ohtsuka M, Quadros RM, Gurumurthy CB. Pronuclear Injection-Based Targeted Transgenesis. CURRENT PROTOCOLS IN HUMAN GENETICS 2016; 91:15.10.1-15.10.28. [PMID: 27727435 PMCID: PMC5123763 DOI: 10.1002/cphg.23] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Microinjection of DNA expression cassettes into fertilized zygotes has been a standard method for generating transgenic animal models. While efficient, the injected DNA integrates randomly into the genome, leading to potential disruption of endogenous genes or regulatory elements, variation in copy number, or integration into heterochromatic regions that inhibit transgene expression. A recently developed method addresses such pitfalls of traditional transgenesis by targeting the transgene to predetermined sites in the genome that can safely harbor exogenous DNA. This method, called Pronuclear Injection-based Targeted Transgenesis (PITT), employs an enzymatic transfer of exogenous DNA from a donor vector to a previously created landing-pad site in the mouse genome. DNA transfer is achieved using molecular tools such as the Cre-LoxP recombinase and PhiC31-attB/P integrase systems. Here, we provide protocols for performing PITT and an overview of the current PITT tools available to the research community. © 2016 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
| | - Masato Ohtsuka
- Department of Molecular Life Science, Division of Basic Medical Science and Molecular Medicine, Tokai University School of Medicine, Isehara, Kanagawa, Japan
- The Institute of Medical Sciences, Tokai University, Isehara, Kanagawa, Japan
| | - Rolen M Quadros
- Mouse Genome Engineering Core Facility, University of Nebraska Medical Center, Omaha, Nebraska
| | - Channabasavaiah B Gurumurthy
- Mouse Genome Engineering Core Facility, University of Nebraska Medical Center, Omaha, Nebraska
- Developmental Neuroscience, Munroe Meyer Institute, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
24
|
Breuss M, Fritz T, Gstrein T, Chan K, Ushakova L, Yu N, Vonberg FW, Werner B, Elling U, Keays DA. Mutations in the murine homologue of TUBB5 cause microcephaly by perturbing cell cycle progression and inducing p53-associated apoptosis. Development 2016; 143:1126-33. [PMID: 26903504 DOI: 10.1242/dev.131516] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 02/15/2016] [Indexed: 12/14/2022]
Abstract
Microtubules play a crucial role in the generation, migration and differentiation of nascent neurons in the developing vertebrate brain. Mutations in the constituents of microtubules, the tubulins, are known to cause an array of neurological disorders, including lissencephaly, polymicrogyria and microcephaly. In this study we explore the genetic and cellular mechanisms that cause TUBB5-associated microcephaly by exploiting two new mouse models: a conditional E401K knock-in, and a conditional knockout animal. These mice present with profound microcephaly due to a loss of upper-layer neurons that correlates with massive apoptosis and upregulation of p53. This phenotype is associated with a delay in cell cycle progression and ectopic DNA elements in progenitors, which is dependent on the dosage of functional Tubb5. Strikingly, we report ectopic Sox2-positive progenitors and defects in spindle orientation in our knock-in mouse line, which are absent in knockout animals. This work sheds light on the functional repertoire of Tubb5, reveals that the E401K mutation acts by a complex mechanism, and demonstrates that the cellular pathology driving TUBB5-associated microcephaly is cell death.
Collapse
Affiliation(s)
- Martin Breuss
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Dr. Bohr-Gasse 7, Vienna 1030, Austria
| | - Tanja Fritz
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Dr. Bohr-Gasse 7, Vienna 1030, Austria
| | - Thomas Gstrein
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Dr. Bohr-Gasse 7, Vienna 1030, Austria
| | - Kelvin Chan
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Dr. Bohr-Gasse 7, Vienna 1030, Austria Medical Scientist Training Program, Stony Brook University Medical Center, Stony Brook, NY 11794, USA
| | - Lyubov Ushakova
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Dr. Bohr-Gasse 7, Vienna 1030, Austria
| | - Nuo Yu
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Dr. Bohr-Gasse 7, Vienna 1030, Austria
| | - Frederick W Vonberg
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Dr. Bohr-Gasse 7, Vienna 1030, Austria Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Barbara Werner
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Dr. Bohr-Gasse 7, Vienna 1030, Austria
| | - Ulrich Elling
- Institute for Molecular Biotechnology (IMBA), Vienna Biocenter (VBC), Dr. Bohr-Gasse 3, Vienna 1030, Austria
| | - David A Keays
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Dr. Bohr-Gasse 7, Vienna 1030, Austria
| |
Collapse
|
25
|
Joyner AL. From Cloning Neural Development Genes to Functional Studies in Mice, 30 Years of Advancements. Curr Top Dev Biol 2016; 116:501-15. [PMID: 26970637 DOI: 10.1016/bs.ctdb.2015.11.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The invention of new mouse molecular genetics techniques, initiated in the 1980s, has repeatedly expanded our ability to tackle exciting developmental biology problems. The brain is the most complex organ, and as such the more sophisticated the molecular genetics technique, the more impact they have on uncovering new insights into how our brain functions. I provide a general time line for the introduction of new techniques over the past 30 years and give examples of new discoveries in the neural development field that emanated from them. I include a look to what the future holds and argue that we are at the dawn of a very exciting age for young scientists interested in studying how the nervous system is constructed and functions with such precision.
Collapse
Affiliation(s)
- Alexandra L Joyner
- Developmental Biology Program, Sloan Kettering Institute, New York, USA.
| |
Collapse
|
26
|
Dodson MV, Allen RE, Du M, Bergen WG, Velleman SG, Poulos SP, Fernyhough-Culver M, Wheeler MB, Duckett SK, Young MRI, Voy BH, Jiang Z, Hausman GJ. INVITED REVIEW: Evolution of meat animal growth research during the past 50 years: Adipose and muscle stem cells. J Anim Sci 2016; 93:457-81. [PMID: 26020737 DOI: 10.2527/jas.2014-8221] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
If one were to compare today's animal growth research to research from a mere 50 yr ago, one would see programs with few similarities. The evolution of this research from whole-animal through cell-based and finally molecular and genomic studies has been enhanced by the identification, isolation, and in vitro evaluation of adipose- and muscle-derived stem cells. This paper will highlight the struggles and the milestones that make this evolving area of research what it is today. The contribution of adipose and muscle stem cell research to development and growth, tissue regeneration, and final carcass composition are reviewed.
Collapse
|
27
|
Suddason T, Anwar S, Charlaftis N, Gallagher E. T-Cell-Specific Deletion of Map3k1 Reveals the Critical Role for Mekk1 and Jnks in Cdkn1b-Dependent Proliferative Expansion. Cell Rep 2016; 14:449-457. [PMID: 26774476 PMCID: PMC4733086 DOI: 10.1016/j.celrep.2015.12.047] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 09/22/2015] [Accepted: 12/07/2015] [Indexed: 12/18/2022] Open
Abstract
MAPK signaling is important for T lymphocyte development, homeostasis, and effector responses. To better understand the role of Mekk1 (encoded by Map3k1) in T cells, we conditionally deleted Map3k1 in LckCre/+Map3k1f/f mice, and these display larger iNKT cell populations within the liver, spleen, and bone marrow. Mekk1 signaling controls splenic and liver iNKT cell expansion in response to glycolipid antigen. LckCre/+Map3k1f/f mice have enhanced liver damage in response to glycolipid antigen. Mekk1 regulates Jnk activation in iNKT cells and binds and transfers Lys63-linked poly-ubiquitin onto Carma1. Map3k1 is critical for the regulation of p27Kip1 (encoded by Cdkn1b). iNKT cell expansion is aberrant in LckCre/+Map3k1f/f mice LckCre/+Map3k1f/f mice have enhanced liver damage in response to glycolipids Mekk1 regulates TCR-dependent Jnk activation Mekk1 regulates p27Kip1 expression to regulate proliferation
Collapse
Affiliation(s)
- Tesha Suddason
- Department of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK.
| | - Saba Anwar
- Department of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Nikolaos Charlaftis
- Department of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Ewen Gallagher
- Department of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK.
| |
Collapse
|
28
|
Genetic engineering of mammals. Cell Tissue Res 2015; 363:289-294. [PMID: 26631228 DOI: 10.1007/s00441-015-2321-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 10/04/2015] [Accepted: 10/27/2015] [Indexed: 01/17/2023]
Abstract
Historically, genetic engineering for mammalian reproductive questions has been accomplished primarily in the mouse. However, all the genetic manipulations that can be done in the mouse can now be accomplished in most domesticated mammals. Random integration of transgenes, homologous recombination and gene editing are now routine for several mammalian species. For livestock, queries related to fertility can be asked directly for the species in question, without a need for a mouse model. For human clinical concerns, the most appropriate model should be selected based on physiology, anatomy, or even size. The mouse will continue to be a useful genetically engineered model. However, other species are now amenable to the full range of genetic manipulations and should be considered as possible models for human conditions when appropriate.
Collapse
|
29
|
Specks J, Nieto-Soler M, Lopez-Contreras AJ, Fernandez-Capetillo O. Modeling the study of DNA damage responses in mice. Methods Mol Biol 2015; 1267:413-37. [PMID: 25636482 DOI: 10.1007/978-1-4939-2297-0_21] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Damaged DNA has a profound impact on mammalian health and overall survival. In addition to being the source of mutations that initiate cancer, the accumulation of toxic amounts of DNA damage can cause severe developmental diseases and accelerate aging. Therefore, understanding how cells respond to DNA damage has become one of the most intense areas of biomedical research in the recent years. However, whereas most mechanistic studies derive from in vitro or in cellulo work, the impact of a given mutation on a living organism is largely unpredictable. For instance, why BRCA1 mutations preferentially lead to breast cancer whereas mutations compromising mismatch repair drive colon cancer is still not understood. In this context, evaluating the specific physiological impact of mutations that compromise genome integrity has become crucial for a better dimensioning of our knowledge. We here describe the various technologies that can be used for modeling mutations in mice and provide a review of the genes and pathways that have been modeled so far in the context of DNA damage responses.
Collapse
Affiliation(s)
- Julia Specks
- Genomic Instability Group, Spanish National Cancer Research Center (CNIO), C/Melchor Fernandez Almagro, 3, E-28029, Madrid, Spain
| | | | | | | |
Collapse
|
30
|
Charlaftis N, Suddason T, Wu X, Anwar S, Karin M, Gallagher E. The MEKK1 PHD ubiquitinates TAB1 to activate MAPKs in response to cytokines. EMBO J 2014; 33:2581-96. [PMID: 25260751 PMCID: PMC4282369 DOI: 10.15252/embj.201488351] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Unlike the other MAP3Ks, MEKK1 (encoded by Map3k1) contains a PHD motif. To understand the role of this motif, we have created a knockin mutant of mouse Map3k1 (Map3k1mPHD) with an inactive PHD motif. Map3k1mPHD ES cells demonstrate that the MEKK1 PHD controls p38 and JNK activation during TGF-β, EGF and microtubule disruption signalling, but does not affect MAPK responses to hyperosmotic stress. Protein microarray profiling identified the adaptor TAB1 as a PHD substrate, and TGF-β- or EGF-stimulated Map3k1mPHD ES cells exhibit defective non-canonical ubiquitination of MEKK1 and TAB1. The MEKK1 PHD binds and mediates the transfer of Lys63-linked poly-Ub, using the conjugating enzyme UBE2N, onto TAB1 to regulate TAK1 and MAPK activation by TGF-β and EGF. Both the MEKK1 PHD and TAB1 are critical for ES-cell differentiation and tumourigenesis. Map3k1mPHD/+ mice exhibit aberrant cardiac tissue, B-cell development, testis and T-cell signalling.
Collapse
Affiliation(s)
| | - Tesha Suddason
- Department of Medicine, Imperial College London, London, UK
| | - Xuefeng Wu
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego School of Medicine, San Diego, CA, USA
| | - Saba Anwar
- Department of Medicine, Imperial College London, London, UK
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego School of Medicine, San Diego, CA, USA
| | - Ewen Gallagher
- Department of Medicine, Imperial College London, London, UK
| |
Collapse
|
31
|
Serra-Hassoun M, Bourgine M, Boniotto M, Berges J, Langa F, Michel ML, Freitas AA, Garcia S. Human hematopoietic reconstitution and HLA-restricted responses in nonpermissive alymphoid mice. THE JOURNAL OF IMMUNOLOGY 2014; 193:1504-11. [PMID: 24973440 DOI: 10.4049/jimmunol.1400412] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We generated a new humanized mouse model to study HLA-restricted immune responses. For this purpose, we created unique murine hosts by enforcing the expression of human SIRPα by murine phagocytes in murine MHC-deficient HLA-transgenic alymphoid hosts, an approach that allowed the immune reconstitution of nonpermissive mice following injection of human hematopoietic stem cells. We showed that these mouse/human chimeras were able to generate HLA-restricted responses to immunization. These new humanized mice may offer attractive models to study immune responses to human diseases, such as HIV and EBV infections, as well as to assay new vaccine strategies.
Collapse
Affiliation(s)
- Malika Serra-Hassoun
- Unité de Biologie des Populations Lymphocytaires, Département d'Immunologie, Institut Pasteur, 75724 Paris, France; Centre National pour la Recherche Scientifique, Unité de Recherche Associée 1961, 75724 Paris, France
| | - Maryline Bourgine
- Laboratoire de Pathogenèse des Virus de l'Hépatite B, Département de Virologie, Institut Pasteur, 75724 Paris, France; INSERM U845, 75724 Paris, France
| | - Michele Boniotto
- Unit of Human Evolutionary Genetics, Department of Genome and Genetics, Institut Pasteur, 75724 Paris, France; and
| | - Julien Berges
- Unité de Biologie des Populations Lymphocytaires, Département d'Immunologie, Institut Pasteur, 75724 Paris, France; Centre National pour la Recherche Scientifique, Unité de Recherche Associée 1961, 75724 Paris, France
| | - Francina Langa
- Centre d'Ingénierie Génétique Murine, Institut Pasteur, 75724 Paris, France
| | - Marie-Louise Michel
- Laboratoire de Pathogenèse des Virus de l'Hépatite B, Département de Virologie, Institut Pasteur, 75724 Paris, France; INSERM U845, 75724 Paris, France
| | - Antonio A Freitas
- Unité de Biologie des Populations Lymphocytaires, Département d'Immunologie, Institut Pasteur, 75724 Paris, France; Centre National pour la Recherche Scientifique, Unité de Recherche Associée 1961, 75724 Paris, France
| | - Sylvie Garcia
- Unité de Biologie des Populations Lymphocytaires, Département d'Immunologie, Institut Pasteur, 75724 Paris, France; Centre National pour la Recherche Scientifique, Unité de Recherche Associée 1961, 75724 Paris, France;
| |
Collapse
|
32
|
Abstract
The protocols in this chapter describe two techniques for the generation of transgenic mice by in vivo manipulation of spermatogonial stem cells (SSCs) with a high rate of success. SSCs in prepubescent animals can either be infected in vivo with recombinant lentiviruses expressing the transgene of interest or DNA can be injected into the testis followed by the application of an electric current resulting in integration of the linearized DNA containing a transgene downstream of the appropriate promoter into SSCs. All male pre-founder mice produced transgenic pups using both protocols with the transgene being heritable. Further, the pre-founder mice could be used in multiple mating experiments resulting in the generation of multiple progeny. These protocols could be extended to perform over-expression/knockdown screens in vivo using bar-coded lentiviruses/plasmid constructs, thus permitting the design of genetic screens in the mouse. Further, these protocols could be adapted to achieve transgenesis in other laboratory animals resulting in the generation of model systems that closely approximate human development and disease.
Collapse
|
33
|
Production of chimeric embryos by aggregation of bovine egfp eight-cell stage blastomeres with two-cell fused and asynchronic embryos. Theriogenology 2013; 80:357-64. [DOI: 10.1016/j.theriogenology.2013.04.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 04/18/2013] [Accepted: 04/19/2013] [Indexed: 01/13/2023]
|
34
|
Abe T, Fujimori T. Reporter mouse lines for fluorescence imaging. Dev Growth Differ 2013; 55:390-405. [PMID: 23621623 DOI: 10.1111/dgd.12062] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 03/19/2013] [Accepted: 03/20/2013] [Indexed: 12/16/2022]
Abstract
The use of live imaging approaches to examine and understand the dynamic processes that take place during mouse development has become widespread. Several groups have reported their success in generating different reporter mouse lines that express a variety of fluorescent markers for imaging. However, there is currently no established database of the reporter mouse lines available for live imaging, such as the Cre transgenic lines (Cre-X-Mice). Researchers therefore often have difficulties in determining which reporter mouse line meets their research purposes. In this review, we summarize some of the reporter mouse lines that have been generated for live imaging studies, and discuss their characteristics.
Collapse
Affiliation(s)
- Takaya Abe
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology (CDB), 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| | | |
Collapse
|
35
|
Goswami M, Lakra WS, Yadav K, Jena JK. Development of an ES-like cell culture system (RESC) from rohu, Labeo rohita (Ham.). FISH PHYSIOLOGY AND BIOCHEMISTRY 2012; 38:1775-1783. [PMID: 22707190 DOI: 10.1007/s10695-012-9674-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Accepted: 05/30/2012] [Indexed: 06/01/2023]
Abstract
An embryonic stem (ES)-like cell culture system RESC from a commercially important freshwater carp, Labeo rohita, was developed using blastula stage embryos. The cells were cultured in Leibovitz-15 (L-15) medium in gelatin-coated cell culture flask supplemented with 15 % fetal bovine serum along with 10 ng ml(-1) basic fibroblast growth factor at 28 °C under feeder-free conditions. The ES-like cells were characterized by their unique morphology, alkaline phosphatase activity, embryoid body formation tendency, expression of transcription factor Oct4, and consistent chromosome count. The RESC cells when treated with retinoic acid differentiated into cells of different lineages. The RESC developed from mid-blastula embryos of L. rohita would be a useful tool for cellular differentiation and gene expression studies.
Collapse
Affiliation(s)
- M Goswami
- National Bureau of Fish Genetic Resources (NBFGR), Lucknow, 226002, UP, India.
| | - W S Lakra
- Central Institute of Fisheries Education, Versova, Andheri (W), Mumbai, 400061, India
| | - Kamalendra Yadav
- National Bureau of Fish Genetic Resources (NBFGR), Lucknow, 226002, UP, India
| | - J K Jena
- National Bureau of Fish Genetic Resources (NBFGR), Lucknow, 226002, UP, India
| |
Collapse
|
36
|
Lee WC, Berry R, Hohenstein P, Davies J. siRNA as a tool for investigating organogenesis: The pitfalls and the promises. Organogenesis 2012; 4:176-81. [PMID: 19279730 DOI: 10.4161/org.4.3.6642] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2007] [Accepted: 03/20/2008] [Indexed: 12/31/2022] Open
Abstract
Removing the function of a specific gene from a developing organ, by making a 'knockout' mouse, is a powerful method for analyzing the molecular pathways that control organogenesis. The technique is expensive, though, in terms of time and money, and complex strategies for producing conditional knockouts are needed for genes that are essential for early development of the embryo, for which an unconditional knockout would be lethal before the organ of interest begins to form. Small interfering RNAs (siRNAs) offer a method of knocking down the expression of specific genes with no need for genomic manipulation. Almost as soon as they had been discovered, siRNAs began to be used to explore the molecular biology of mammalian cells in conventional, two-dimensional culture. They have now also been applied successfully, by several groups, to knock down specific genes in various organ rudiments developing in organ culture. This article reviews the basic technique of siRNA-mediated gene knockdown and how it is being applied to organ culture. It also reviews some of the current problems and challenges in the field, and the ways in which these problems are likely to be overcome.
Collapse
Affiliation(s)
- Wen-Chin Lee
- Centre for Integrative Physiology; University of Edinburgh; Edinburgh UK
| | | | | | | |
Collapse
|
37
|
Abstract
Mouse genetic engineering has revolutionized our understanding of the molecular and genetic basis of heart development and disease. This technology involves conditional tissue-specific and temporal transgenic and gene targeting approaches, as well as introduction of polymorphisms into the mouse genome. These approaches are increasingly used to elucidate the genetic pathways underlying tissue homeostasis, physiology, and pathophysiology of adult heart. They have also led to the development of clinically relevant models of human cardiac diseases. Here, we review the technologies and their limitations in general and the cardiovascular research community in particular.
Collapse
Affiliation(s)
- Thomas Doetschman
- BIO5 Institute and Department of Cellular & Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | | |
Collapse
|
38
|
Kawai N, Ochiai H, Sakuma T, Yamada L, Sawada H, Yamamoto T, Sasakura Y. Efficient targeted mutagenesis of the chordate Ciona intestinalis genome with zinc-finger nucleases. Dev Growth Differ 2012; 54:535-45. [PMID: 22640377 DOI: 10.1111/j.1440-169x.2012.01355.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Revised: 04/06/2012] [Accepted: 04/09/2012] [Indexed: 12/13/2022]
Abstract
Zinc-finger nucleases (ZFNs) are engineered nucleases that induce DNA double-strand breaks (DSBs) at target sequences. They have been used as tools for generating targeted mutations in the genomes of multiple organisms in both animals and plants. The DSB induced by ZFNs is repaired by non-homologous end joining (NHEJ) or by homologous recombination (HR) mechanisms. Non-homologous end joining induces some errors because it is independent of a reference DNA sequence. Through the NHEJ mechanism, ZFNs generate insertional or deletional mutations at the target sequence. We examined the usability, specificity and toxicity of ZFNs in the basal chordate Ciona intestinalis. As the target of ZFNs, we chose an enhanced green fluorescent protein (EGFP) gene artificially inserted in the C. intestinalis genome because this locus is neutral for the development and growth of C. intestinalis, and the efficiency of mutagenesis with ZFNs can thus be determined without any bias. We introduced EGFP -ZFN mRNAs into the embryos of an EGFP -transgenic line and observed the mutation frequency in the target site of EGFP . We also examined the effects of the EGFP -ZFNs at off-target sites resembling the EGFP target sequence in the C. intestinalis genome in order to examine the specificity of ZFNs. We further investigated the influence of ZFNs on embryogenesis, and showed that adequate amounts of ZFNs, which do not disrupt embryogenesis, can efficiently induce mutations on the on-target site with less effect on the off-target sites. This suggests that target mutagenesis with ZFNs will be a powerful technique in C. intestinalis.
Collapse
Affiliation(s)
- Narudo Kawai
- Shimoda Marine Research Center, University of Tsukuba, Shimoda, Shizuoka, 415-0025, Japan.
| | | | | | | | | | | | | |
Collapse
|
39
|
Howell JC, Wells JM. Generating intestinal tissue from stem cells: potential for research and therapy. Regen Med 2012; 6:743-55. [PMID: 22050526 DOI: 10.2217/rme.11.90] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Intestinal resection and malformations in adult and pediatric patients result in devastating consequences. Unfortunately, allogeneic transplantation of intestinal tissue into patients has not been met with the same measure of success as the transplantation of other organs. Attempts to engineer intestinal tissue in vitro include disaggregation of adult rat intestine into subunits called organoids, harvesting native adult stem cells from mouse intestine and spontaneous generation of intestinal tissue from embryoid bodies. Recently, by utilizing principles gained from the study of developmental biology, human pluripotent stem cells have been demonstrated to be capable of directed differentiation into intestinal tissue in vitro. Pluripotent stem cells offer a unique and promising means to generate intestinal tissue for the purposes of modeling intestinal disease, understanding embryonic development and providing a source of material for therapeutic transplantation.
Collapse
Affiliation(s)
- Jonathan C Howell
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229-3039, USA
| | | |
Collapse
|
40
|
Nanoinjection: pronuclear DNA delivery using a charged lance. Transgenic Res 2012; 21:1279-90. [PMID: 22415347 DOI: 10.1007/s11248-012-9610-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Accepted: 03/02/2012] [Indexed: 02/06/2023]
Abstract
We present a non-fluidic pronuclear injection method using a silicon microchip "nanoinjector" composed of a microelectromechanical system with a solid, electrically conductive lance. Unlike microinjection which uses fluid delivery of DNA, nanoinjection electrically accumulates DNA on the lance, the DNA-coated lance is inserted into the pronucleus, and DNA is electrically released. We compared nanoinjection and microinjection side-by-side over the course of 4 days, injecting 1,013 eggs between the two groups. Nanoinjected zygotes had significantly higher rates of integration per injected embryo, with 6.2% integration for nanoinjected embryos compared to 1.6% integration for microinjected embryos. This advantage is explained by nanoinjected zygotes' significantly higher viability in two stages of development: zygote progress to two-cell stage, and progress from two-cell stage embryos to birth. We observed that 77.6% of nanoinjected zygotes proceeded to two-cell stage compared to 54.7% of microinjected zygotes. Of the healthy two-cell stage embryos, 52.4% from the nanoinjection group and 23.9% from the microinjected group developed into pups. Structural advantages of the nanoinjector are likely to contribute to the high viability observed. For instance, because charge is used to retain and release DNA, extracellular fluid is not injected into the pronucleus and the cross-sectional area of the nanoinjection lance (0.06 µm(2)) is smaller than that of a microinjection pipette tip (0.78 µm(2)). According to results from the comparative nanoinjection versus microinjection study, we conclude that nanoinjection is a viable method of pronuclear DNA transfer which presents viability advantages over microinjection.
Collapse
|
41
|
Sehgal L, Thorat R, Khapare N, Mukhopadhaya A, Sawant M, Dalal SN. Lentiviral mediated transgenesis by in vivo manipulation of spermatogonial stem cells. PLoS One 2011; 6:e21975. [PMID: 21760937 PMCID: PMC3131306 DOI: 10.1371/journal.pone.0021975] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2011] [Accepted: 06/14/2011] [Indexed: 11/19/2022] Open
Abstract
This report describes a technique for the generation of transgenic mice by in vivo manipulation of spermatogonial stem cells with a high rate of success. Spermatogonial stem cells (SSCs) in pre-pubescent animals were infected in vivo with recombinant lentiviruses expressing EGFP-f and mated with normal females. All male pre-founder mice produced transgenic pups with an overall success rate of over 60%. The transgene was heritable and the pre-founder mice could be used in multiple mating experiments. This technology could be used to perform overexpression/knockdown screens in vivo using bar-coded lentiviruses, thus permitting the design of genetic screens in the mouse. Further, this technology could be adapted to other laboratory animals resulting in the generation of model systems that closely approximate human development and disease.
Collapse
Affiliation(s)
- Lalit Sehgal
- KS-215, Advanced Centre for Treatment Research Education and Cancer, Tata Memorial Centre, Kharghar Node, Navi Mumbai, India
| | - Rahul Thorat
- KS-215, Advanced Centre for Treatment Research Education and Cancer, Tata Memorial Centre, Kharghar Node, Navi Mumbai, India
| | - Nileema Khapare
- KS-215, Advanced Centre for Treatment Research Education and Cancer, Tata Memorial Centre, Kharghar Node, Navi Mumbai, India
| | - Amitabha Mukhopadhaya
- KS-215, Advanced Centre for Treatment Research Education and Cancer, Tata Memorial Centre, Kharghar Node, Navi Mumbai, India
| | - Mugdha Sawant
- KS-215, Advanced Centre for Treatment Research Education and Cancer, Tata Memorial Centre, Kharghar Node, Navi Mumbai, India
| | - Sorab N. Dalal
- KS-215, Advanced Centre for Treatment Research Education and Cancer, Tata Memorial Centre, Kharghar Node, Navi Mumbai, India
- * E-mail:
| |
Collapse
|
42
|
Jones D. Genetic engineering of a mouse: Dr. Frank Ruddle and somatic cell genetics. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2011; 84:117-24. [PMID: 21698043 PMCID: PMC3117405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Genetic engineering is the process of modifying an organism's genetic composition by adding foreign genes to produce desired traits or evaluate function. Dr. Jon W. Gordon and Sterling Professor Emeritus at Yale Dr. Frank H. Ruddle were pioneers in mammalian gene transfer research. Their research resulted in production of the first transgenic animals, which contained foreign DNA that was passed on to offspring. Transgenic mice have revolutionized biology, medicine, and biotechnology in the 21st century. In brief, this review revisits their creation of transgenic mice and discusses a few evolving applications of their transgenic technology used in biomedical research.
Collapse
|
43
|
Affiliation(s)
- THOMAS DOETSCHMAN
- BIO5 Institute and Department of Cell Biology and Anatomy, University of Arizona, Tucson, Arizona
| |
Collapse
|
44
|
Bressan FF, Dos Santos Miranda M, Perecin F, De Bem TH, Pereira FTV, Russo-Carbolante EM, Alves D, Strauss B, Bajgelman M, Krieger JE, Binelli M, Meirelles FV. Improved production of genetically modified fetuses with homogeneous transgene expression after transgene integration site analysis and recloning in cattle. Cell Reprogram 2011; 13:29-36. [PMID: 21241190 DOI: 10.1089/cell.2010.0022] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Animal cloning by nuclear transfer (NT) has made the production of transgenic animals using genetically modified donor cells possible and ensures the presence of the gene construct in the offspring. The identification of transgene insertion sites in donor cells before cloning may avoid the production of animals that carry undesirable characteristics due to positional effects. This article compares blastocyst development and competence to establish pregnancies of bovine cloned embryos reconstructed with lentivirus-mediated transgenic fibroblasts containing either random integration of a transgene (random integration group) or nuclear transfer derived transgenic fibroblasts with known transgene insertion sites submitted to recloning (recloned group). In the random integration group, eGFP-expressing bovine fetal fibroblasts were selected by fluorescence activated cell sorting (FACS) and used as nuclei donor cells for NT. In the recloned group, a fibroblast cell line derived from a transgenic cloned fetus was characterized regarding transgene insertion and submitted to recloning. The recloned group had higher blastocyst production (25.38 vs. 14.42%) and higher percentage of 30-day pregnancies (14.29 vs. 2.56%) when compared to the random integration group. Relative eGFP expression analysis in fibroblasts derived from each cloned embryo revealed more homogeneous expression in the recloned group. In conclusion, the use of cell lines recovered from transgenic fetuses after identification of the transgene integration site allowed for the production of cells and fetuses with stable transgene expression, and recloning may improve transgenic animal yields.
Collapse
|
45
|
Katsumoto K, Shiraki N, Miki R, Kume S. Embryonic and adult stem cell systems in mammals: ontology and regulation. Dev Growth Differ 2010; 52:115-29. [PMID: 20078654 DOI: 10.1111/j.1440-169x.2009.01160.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Stem cells are defined as having the ability to self-renew and to generate differentiated cells. During embryogenesis, cells are initially proliferative and pluripotent and then they gradually become restricted to different cell fates. In the adult, tissue stem cells are normally quiescent, but become proliferative upon injury. Knowledge from developmental biology and insights into the properties of stem cells are keys to further understanding and successful manipulation. Here, we first focus on ES cells, then on embryonic development, and then on tissue stem cells of endodermally derived tissues, particularly the liver and pancreas.
Collapse
Affiliation(s)
- Keiichi Katsumoto
- Department of Stem Cell Biology, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Honjo 2-2-1, Kumamoto 860-0811, Japan
| | | | | | | |
Collapse
|
46
|
Abstract
Experimental mouse chimeras have served as immensely important research tools for studying many aspects of mammalian development ever since they first were produced over 50 years ago. Chimera studies have served as crucial assays in the era of modern mouse genetics that was triggered by the advent of mouse embryonic stem cells. Lately, chimeras are also used as proof of pluripotency and normality of induced pluripotent stem cells. With this long history in mind, it may seem surprising that chimeras now have an ever-increasing role to play. The high-throughput mouse gene targeting projects are in the process of producing ES cell lines with a mutation in each of the close to 20,000 known protein coding genes. These will all be waiting for germline transmission through chimeras. Such a large-scale approach calls for simplified methods for generating germline transmitting chimeras. In this chapter, we will describe the currently most cost efficient and simple method; the aggregation of pluripotent stem cells with diploid or tetraploid mouse embryos. Since most of the large knockout projects are using the C57BL/6 background, we will pay special attention to cell lines derived from this inbred strain.
Collapse
|
47
|
Limaye A, Hall B, Kulkarni AB. Manipulation of mouse embryonic stem cells for knockout mouse production. ACTA ACUST UNITED AC 2009; Chapter 19:Unit 19.13 19.13.1-24. [PMID: 19731225 DOI: 10.1002/0471143030.cb1913s44] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The establishment of mouse embryonic stem (ES) cell lines has allowed for the gene?ration of the knockout mouse. ES cells that are genetically altered in culture can then be manipulated to derive a whole mouse containing the desired mutation. To successfully generate a knockout mouse, however, the ES cells must be carefully cultivated in a pluripotent state throughout the gene-targeting experiment. This unit describes detailed step-by-step protocols, reagents, equipment, and strategies needed for the successful generation of gene knockout embryonic stem cells using homologous recombination technologies.
Collapse
Affiliation(s)
- Advait Limaye
- Department of Health and Human Services, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | | | | |
Collapse
|
48
|
Bressan FF, De Bem THC, Perecin F, Lopes FL, Ambrosio CE, Meirelles FV, Miglino MA. Unearthing the roles of imprinted genes in the placenta. Placenta 2009; 30:823-34. [PMID: 19679348 DOI: 10.1016/j.placenta.2009.07.007] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2009] [Accepted: 07/22/2009] [Indexed: 11/17/2022]
Abstract
Mammalian fetal survival and growth are dependent on a well-established and functional placenta. Although transient, the placenta is the first organ to be formed during pregnancy and is responsible for important functions during development, such as the control of metabolism and fetal nutrition, gas and metabolite exchange, and endocrine control. Epigenetic marks and gene expression patterns in early development play an essential role in embryo and fetal development. Specifically, the epigenetic phenomenon known as genomic imprinting, represented by the non-equivalence of the paternal and maternal genome, may be one of the most important regulatory pathways involved in the development and function of the placenta in eutherian mammals. A lack of pattern or an imprecise pattern of genomic imprinting can lead to either embryonic losses or a disruption in fetal and placental development. Genetically modified animals present a powerful approach for revealing the interplay between gene expression and placental function in vivo and allow a single gene disruption to be analyzed, particularly focusing on its role in placenta function. In this paper, we review the recent transgenic strategies that have been successfully created in order to provide a better understanding of the epigenetic patterns of the placenta, with a special focus on imprinted genes. We summarize a number of phenotypes derived from the genetic manipulation of imprinted genes and other epigenetic modulators in an attempt to demonstrate that gene-targeting studies have contributed considerably to the knowledge of placentation and conceptus development.
Collapse
Affiliation(s)
- F F Bressan
- Department of Basic Sciences, Faculty of Animal Sciences and Food Engineering, University of São Paulo, Pirassununga, Brazil
| | | | | | | | | | | | | |
Collapse
|
49
|
Bratt-Leal AM, Carpenedo RL, McDevitt TC. Engineering the embryoid body microenvironment to direct embryonic stem cell differentiation. Biotechnol Prog 2009; 25:43-51. [PMID: 19198003 DOI: 10.1002/btpr.139] [Citation(s) in RCA: 213] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Embryonic stem cells (ESCs) are pluripotent cells capable of differentiating into all somatic and germ cell types. The intrinsic ability of pluripotent cells to generate a vast array of different cells makes ESCs a robust resource for a variety of cell transplantation and tissue engineering applications, however, efficient and controlled means of directing ESC differentiation is essential for the development of regenerative therapies. ESCs are commonly differentiated in vitro by spontaneously self-assembling in suspension culture into 3D cell aggregates called embryoid bodies (EBs), which mimic many of the hallmarks of early embryonic development, yet the 3D organization and structure of EBs also presents unique challenges to effectively direct the differentiation of the cells. ESC differentiation is strongly influenced by physical and chemical signals comprising the local extracellular microenvironment, thus current methods to engineer EB differentiation have focused primarily on spatially controlling EB size, adding soluble factors to the media, or culturing EBs on or within natural or synthetic extracellular matrices. Although most such strategies aim to influence differentiation from the exterior of EBs, engineering the microenvironment directly within EBs enables new opportunities to efficiently direct the fate of the cells by locally controlling the presentation of morphogenic cues.
Collapse
Affiliation(s)
- Andrés M Bratt-Leal
- The Wallace H. Coulter Dept. of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, GA, USA
| | | | | |
Collapse
|
50
|
Abstract
In this chapter we give an overview of mutagenesis methods in the mouse as they evolved over the last two decades, an outlook of ongoing and future developments and advice for choosing a mutagenesis strategy. Where appropriate, reference is given to relevant chapters of this book, key original articles and links of web-based resources for mouse mutagenesis.
Collapse
Affiliation(s)
- Ralf Kühn
- Institute for Developmental Genetics, Helmholtz Zentrum München - German Research Center for Environmental Health, Munich, Germany
| | | |
Collapse
|