1
|
Liang W, He J, Mao C, Yu C, Meng Q, Xue J, Wu X, Li S, Wang Y, Yi H. Gene editing monkeys: Retrospect and outlook. Front Cell Dev Biol 2022; 10:913996. [PMID: 36158194 PMCID: PMC9493099 DOI: 10.3389/fcell.2022.913996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
Animal models play a key role in life science research, especially in the study of human disease pathogenesis and drug screening. Because of the closer proximity to humans in terms of genetic evolution, physiology, immunology, biochemistry, and pathology, nonhuman primates (NHPs) have outstanding advantages in model construction for disease mechanism study and drug development. In terms of animal model construction, gene editing technology has been widely applied to this area in recent years. This review summarizes the current progress in the establishment of NHPs using gene editing technology, which mainly focuses on rhesus and cynomolgus monkeys. In addition, we discuss the limiting factors in the applications of genetically modified NHP models as well as the possible solutions and improvements. Furthermore, we highlight the prospects and challenges of the gene-edited NHP models.
Collapse
Affiliation(s)
- Weizheng Liang
- Central Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
- *Correspondence: Weizheng Liang, ; Shanliang Li, ; Yukai Wang, ; Hongyang Yi,
| | - Junli He
- Department of Pediatrics, Shenzhen University General Hospital, Shenzhen, China
| | - Chenyu Mao
- University of Pennsylvania, Philadelphia, PA, United States
| | - Chengwei Yu
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Qingxue Meng
- Central Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Jun Xue
- Department of General Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Xueliang Wu
- Department of General Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Shanliang Li
- Department of Pharmacology, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- *Correspondence: Weizheng Liang, ; Shanliang Li, ; Yukai Wang, ; Hongyang Yi,
| | - Yukai Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, China
- *Correspondence: Weizheng Liang, ; Shanliang Li, ; Yukai Wang, ; Hongyang Yi,
| | - Hongyang Yi
- National Clinical Research Centre for Infectious Diseases, The Third People’s Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
- *Correspondence: Weizheng Liang, ; Shanliang Li, ; Yukai Wang, ; Hongyang Yi,
| |
Collapse
|
2
|
Yoshimatsu S, Seki F, Okahara J, Watanabe H, Sasaguri H, Haga Y, Hata JI, Sanosaka T, Inoue T, Mineshige T, Lee CY, Shinohara H, Kurotaki Y, Komaki Y, Kishi N, Murayama AY, Nagai Y, Minamimoto T, Yamamoto M, Nakajima M, Zhou Z, Nemoto A, Sato T, Ikeuchi T, Sahara N, Morimoto S, Shiozawa S, Saido TC, Sasaki E, Okano H. Multimodal analyses of a non-human primate model harboring mutant amyloid precursor protein transgenes driven by the human EF1α promoter. Neurosci Res 2022; 185:49-61. [PMID: 36075457 DOI: 10.1016/j.neures.2022.08.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 08/18/2022] [Accepted: 08/21/2022] [Indexed: 11/30/2022]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia which afflicts tens of millions of people worldwide. Despite many scientific progresses to dissect the AD's molecular basis from studies on various mouse models, it has been suffered from evolutionary species differences. Here, we report generation of a non-human primate (NHP), common marmoset model ubiquitously expressing Amyloid-beta precursor protein (APP) transgenes with the Swedish (KM670/671NL) and Indiana (V717F) mutations. The transgene integration of generated two transgenic marmosets (TG1&TG2) was thoroughly investigated by genomic PCR, whole-genome sequencing, and fluorescence in situ hybridization. By reprogramming, we confirmed the validity of transgene expression in induced neurons in vitro. Moreover, we discovered structural changes in specific brain regions of transgenic marmosets by magnetic resonance imaging analysis, including in the entorhinal cortex and hippocampus. In immunohistochemistry, we detected increased Aβ plaque-like structures in TG1 brain at 7 years old, although evident neuronal loss or glial inflammation was not observed. Thus, this study summarizes our attempt to establish an NHP AD model. Although the transgenesis approach alone seemed not sufficient to fully recapitulate AD in NHPs, it may be beneficial for drug development and further disease modeling by combination with other genetically engineered models and disease-inducing approaches.
Collapse
Affiliation(s)
- Sho Yoshimatsu
- Department of Physiology, School of Medicine, Keio University, Shinjuku-ku, Tokyo 160-8582, Japan; Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako City, Saitama 351-0198, Japan; Laboratory for Marmoset Neural Architecture, RIKEN Center for Brain Science, Wako City, Saitama 351-0198, Japan
| | - Fumiko Seki
- Department of Physiology, School of Medicine, Keio University, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Junko Okahara
- Laboratory for Marmoset Neural Architecture, RIKEN Center for Brain Science, Wako City, Saitama 351-0198, Japan
| | - Hirotaka Watanabe
- Department of Physiology, School of Medicine, Keio University, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hiroki Sasaguri
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako City, Saitama 351-0198, Japan
| | - Yawara Haga
- Laboratory for Marmoset Neural Architecture, RIKEN Center for Brain Science, Wako City, Saitama 351-0198, Japan; Graduate School of Human Health Sciences, Tokyo Metropolitan University, Arakawa-ku, Tokyo 116-8551, Japan
| | - Jun-Ichi Hata
- Department of Physiology, School of Medicine, Keio University, Shinjuku-ku, Tokyo 160-8582, Japan; Laboratory for Marmoset Neural Architecture, RIKEN Center for Brain Science, Wako City, Saitama 351-0198, Japan; Graduate School of Human Health Sciences, Tokyo Metropolitan University, Arakawa-ku, Tokyo 116-8551, Japan
| | - Tsukasa Sanosaka
- Department of Physiology, School of Medicine, Keio University, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Takashi Inoue
- Department of Marmoset Biology and Medicine, Central Institute for Experimental Animals, Kawasaki, Kanagawa 210-0821, Japan
| | - Takayuki Mineshige
- Department of Marmoset Biology and Medicine, Central Institute for Experimental Animals, Kawasaki, Kanagawa 210-0821, Japan
| | - Chia-Ying Lee
- Department of Marmoset Biology and Medicine, Central Institute for Experimental Animals, Kawasaki, Kanagawa 210-0821, Japan
| | - Haruka Shinohara
- Department of Marmoset Biology and Medicine, Central Institute for Experimental Animals, Kawasaki, Kanagawa 210-0821, Japan
| | - Yoko Kurotaki
- Department of Marmoset Biology and Medicine, Central Institute for Experimental Animals, Kawasaki, Kanagawa 210-0821, Japan
| | - Yuji Komaki
- Live Imaging Center, Central Institute for Experimental Animals, Kawasaki, Kanagawa 210-0821, Japan
| | - Noriyuki Kishi
- Department of Physiology, School of Medicine, Keio University, Shinjuku-ku, Tokyo 160-8582, Japan; Laboratory for Marmoset Neural Architecture, RIKEN Center for Brain Science, Wako City, Saitama 351-0198, Japan
| | - Ayaka Y Murayama
- Department of Physiology, School of Medicine, Keio University, Shinjuku-ku, Tokyo 160-8582, Japan; Laboratory for Marmoset Neural Architecture, RIKEN Center for Brain Science, Wako City, Saitama 351-0198, Japan
| | - Yuji Nagai
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba City, Chiba 263-8555, Japan
| | - Takafumi Minamimoto
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba City, Chiba 263-8555, Japan
| | - Masafumi Yamamoto
- ICLAS Monitoring Center, Central Institute for Experimental Animals, Kanagawa 210-0821, Japan
| | - Mayutaka Nakajima
- Department of Physiology, School of Medicine, Keio University, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Zhi Zhou
- Department of Physiology, School of Medicine, Keio University, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Akisa Nemoto
- Department of Physiology, School of Medicine, Keio University, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Tsukika Sato
- Department of Physiology, School of Medicine, Keio University, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Chuo-ku, Niigata 951-8122, Japan
| | - Naruhiko Sahara
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba City, Chiba 263-8555, Japan
| | - Satoru Morimoto
- Department of Physiology, School of Medicine, Keio University, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Seiji Shiozawa
- Department of Physiology, School of Medicine, Keio University, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako City, Saitama 351-0198, Japan
| | - Erika Sasaki
- Laboratory for Marmoset Neural Architecture, RIKEN Center for Brain Science, Wako City, Saitama 351-0198, Japan; Department of Marmoset Biology and Medicine, Central Institute for Experimental Animals, Kawasaki, Kanagawa 210-0821, Japan.
| | - Hideyuki Okano
- Department of Physiology, School of Medicine, Keio University, Shinjuku-ku, Tokyo 160-8582, Japan; Laboratory for Marmoset Neural Architecture, RIKEN Center for Brain Science, Wako City, Saitama 351-0198, Japan.
| |
Collapse
|
3
|
Schmidt JK, Reynolds MR, Golos TG, Slukvin II. CRISPR/Cas9 genome editing to create nonhuman primate models for studying stem cell therapies for HIV infection. Retrovirology 2022; 19:17. [PMID: 35948929 PMCID: PMC9363854 DOI: 10.1186/s12977-022-00604-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 07/28/2022] [Indexed: 12/13/2022] Open
Abstract
Nonhuman primates (NHPs) are well-established basic and translational research models for human immunodeficiency virus (HIV) infections and pathophysiology, hematopoietic stem cell (HSC) transplantation, and assisted reproductive technologies. Recent advances in CRISPR/Cas9 gene editing technologies present opportunities to refine NHP HIV models for investigating genetic factors that affect HIV replication and designing cellular therapies that exploit genetic barriers to HIV infections, including engineering mutations into CCR5 and conferring resistance to HIV/simian immunodeficiency virus (SIV) infections. In this report, we provide an overview of recent advances and challenges in gene editing NHP embryos and discuss the value of genetically engineered animal models for developing novel stem cell-based therapies for curing HIV.
Collapse
Affiliation(s)
- Jenna Kropp Schmidt
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Matthew R Reynolds
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Thaddeus G Golos
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, USA
| | - Igor I Slukvin
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Pathology and Laboratory Medicine, Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1220 Capitol Court, Madison, WI, 53715, USA.
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
4
|
Schmidt JK, Jones KM, Van Vleck T, Emborg ME. Modeling genetic diseases in nonhuman primates through embryonic and germline modification: Considerations and challenges. Sci Transl Med 2022; 14:eabf4879. [PMID: 35235338 PMCID: PMC9373237 DOI: 10.1126/scitranslmed.abf4879] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Genetic modification of the embryo or germ line of nonhuman primates is envisioned as a method to develop improved models of human disease, yet the promise of such animal models remains unfulfilled. Here, we discuss current methods and their limitations for producing nonhuman primate genetic models that faithfully genocopy and phenocopy human disease. We reflect on how to ethically maximize the translational relevance of such models in the search for new therapeutic strategies to treat human disease.
Collapse
Affiliation(s)
- Jenna K. Schmidt
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Kathryn M. Jones
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Trevor Van Vleck
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Marina E. Emborg
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
5
|
Rodriguez-Polo I, Mißbach S, Petkov S, Mattern F, Maierhofer A, Grządzielewska I, Tereshchenko Y, Urrutia-Cabrera D, Haaf T, Dressel R, Bartels I, Behr R. A piggyBac-based platform for genome editing and clonal rhesus macaque iPSC line derivation. Sci Rep 2021; 11:15439. [PMID: 34326359 PMCID: PMC8322147 DOI: 10.1038/s41598-021-94419-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 07/05/2021] [Indexed: 02/07/2023] Open
Abstract
Non-human primates (NHPs) are, due to their close phylogenetic relationship to humans, excellent animal models to study clinically relevant mutations. However, the toolbox for the genetic modification of NHPs is less developed than those for other species like mice. Therefore, it is necessary to further develop and refine genome editing approaches in NHPs. NHP pluripotent stem cells (PSCs) share key molecular signatures with the early embryo, which is an important target for genomic modification. Therefore, PSCs are a valuable test system for the validation of embryonic genome editing approaches. In the present study, we made use of the versatility of the piggyBac transposon system for different purposes in the context of NHP stem cell technology and genome editing. These include (1) Robust reprogramming of rhesus macaque fibroblasts to induced pluripotent stem cells (iPSCs); (2) Culture of the iPSCs under feeder-free conditions even after removal of the transgene resulting in transgene-free iPSCs; (3) Development of a CRISPR/Cas-based work-flow to edit the genome of rhesus macaque PSCs with high efficiency; (4) Establishment of a novel protocol for the derivation of gene-edited monoclonal NHP-iPSC lines. These findings facilitate efficient testing of genome editing approaches in NHP-PSC before their in vivo application.
Collapse
Affiliation(s)
- Ignacio Rodriguez-Polo
- Research Platform Degenerative Diseases, German Primate Center-Leibniz Institute for Primate Research, Kellnerweg 4, 37077, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Göttingen, Göttingen, Germany
| | - Sophie Mißbach
- Research Platform Degenerative Diseases, German Primate Center-Leibniz Institute for Primate Research, Kellnerweg 4, 37077, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Göttingen, Göttingen, Germany
| | - Stoyan Petkov
- Research Platform Degenerative Diseases, German Primate Center-Leibniz Institute for Primate Research, Kellnerweg 4, 37077, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Göttingen, Göttingen, Germany
| | - Felix Mattern
- Institut für Humangenetik, Universität Würzburg, Biozentrum, Am Hubland, 97074, Würzburg, Germany
| | - Anna Maierhofer
- Institut für Humangenetik, Universität Würzburg, Biozentrum, Am Hubland, 97074, Würzburg, Germany
| | - Iga Grządzielewska
- Research Platform Degenerative Diseases, German Primate Center-Leibniz Institute for Primate Research, Kellnerweg 4, 37077, Göttingen, Germany
- Max Planck Molecular Biology Program (M.Sc./Ph.D.), Justus-von-Liebig-Weg 11, 37077, Göttingen, Germany
| | - Yuliia Tereshchenko
- Research Platform Degenerative Diseases, German Primate Center-Leibniz Institute for Primate Research, Kellnerweg 4, 37077, Göttingen, Germany
- Max Planck Molecular Biology Program (M.Sc./Ph.D.), Justus-von-Liebig-Weg 11, 37077, Göttingen, Germany
| | - Daniel Urrutia-Cabrera
- Research Platform Degenerative Diseases, German Primate Center-Leibniz Institute for Primate Research, Kellnerweg 4, 37077, Göttingen, Germany
- Cellular Reprogramming Unit, Center for Eye Research Australia, 75 Commercial Road, Melbourne, 3004, Australia
| | - Thomas Haaf
- Institut für Humangenetik, Universität Würzburg, Biozentrum, Am Hubland, 97074, Würzburg, Germany
| | - Ralf Dressel
- German Center for Cardiovascular Research (DZHK), Partner site Göttingen, Göttingen, Germany
- Institute of Cellular and Molecular Immunology, University Medical Center Göttingen, Humboldtalle 34, 37073, Göttingen, Germany
| | - Iris Bartels
- Institute of Human Genetics, University Medical Center Göttingen, Robert-Koch-Str. 40, 37075, Göttingen, Germany
| | - Rüdiger Behr
- Research Platform Degenerative Diseases, German Primate Center-Leibniz Institute for Primate Research, Kellnerweg 4, 37077, Göttingen, Germany.
- German Center for Cardiovascular Research (DZHK), Partner site Göttingen, Göttingen, Germany.
| |
Collapse
|
6
|
Holtze S, Gorshkova E, Braude S, Cellerino A, Dammann P, Hildebrandt TB, Hoeflich A, Hoffmann S, Koch P, Terzibasi Tozzini E, Skulachev M, Skulachev VP, Sahm A. Alternative Animal Models of Aging Research. Front Mol Biosci 2021; 8:660959. [PMID: 34079817 PMCID: PMC8166319 DOI: 10.3389/fmolb.2021.660959] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 04/08/2021] [Indexed: 12/23/2022] Open
Abstract
Most research on mechanisms of aging is being conducted in a very limited number of classical model species, i.e., laboratory mouse (Mus musculus), rat (Rattus norvegicus domestica), the common fruit fly (Drosophila melanogaster) and roundworm (Caenorhabditis elegans). The obvious advantages of using these models are access to resources such as strains with known genetic properties, high-quality genomic and transcriptomic sequencing data, versatile experimental manipulation capabilities including well-established genome editing tools, as well as extensive experience in husbandry. However, this approach may introduce interpretation biases due to the specific characteristics of the investigated species, which may lead to inappropriate, or even false, generalization. For example, it is still unclear to what extent knowledge of aging mechanisms gained in short-lived model organisms is transferable to long-lived species such as humans. In addition, other specific adaptations favoring a long and healthy life from the immense evolutionary toolbox may be entirely missed. In this review, we summarize the specific characteristics of emerging animal models that have attracted the attention of gerontologists, we provide an overview of the available data and resources related to these models, and we summarize important insights gained from them in recent years. The models presented include short-lived ones such as killifish (Nothobranchius furzeri), long-lived ones such as primates (Callithrix jacchus, Cebus imitator, Macaca mulatta), bathyergid mole-rats (Heterocephalus glaber, Fukomys spp.), bats (Myotis spp.), birds, olms (Proteus anguinus), turtles, greenland sharks, bivalves (Arctica islandica), and potentially non-aging ones such as Hydra and Planaria.
Collapse
Affiliation(s)
- Susanne Holtze
- Department of Reproduction Management, Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany
| | - Ekaterina Gorshkova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Stan Braude
- Department of Biology, Washington University in St. Louis, St. Louis, MO, United States
| | - Alessandro Cellerino
- Biology Laboratory, Scuola Normale Superiore, Pisa, Italy
- Leibniz Institute on Aging – Fritz Lipmann Institute, Jena, Germany
| | - Philip Dammann
- Department of General Zoology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
- Central Animal Laboratory, University Hospital Essen, Essen, Germany
| | - Thomas B. Hildebrandt
- Department of Reproduction Management, Leibniz Institute for Zoo and Wildlife Research, Berlin, Germany
- Faculty of Veterinary Medicine, Free University of Berlin, Berlin, Germany
| | - Andreas Hoeflich
- Division Signal Transduction, Institute for Genome Biology, Leibniz Institute for Farm Animal Biology, Dummerstorf, Germany
| | - Steve Hoffmann
- Computational Biology Group, Leibniz Institute on Aging – Fritz Lipmann Institute, Jena, Germany
| | - Philipp Koch
- Core Facility Life Science Computing, Leibniz Institute on Aging – Fritz Lipmann Institute, Jena, Germany
| | - Eva Terzibasi Tozzini
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Naples, Italy
| | - Maxim Skulachev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Vladimir P. Skulachev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Arne Sahm
- Computational Biology Group, Leibniz Institute on Aging – Fritz Lipmann Institute, Jena, Germany
| |
Collapse
|
7
|
Schmidt JK, Mean KD, Dusek BM, Hinkle HM, Puntney RC, Alexander ES, Malicki KB, Sneed EL, Moy AW, Golos TG. Comparative computer-assisted sperm analysis in non-human primates. J Med Primatol 2021; 50:108-119. [PMID: 33469948 PMCID: PMC7969417 DOI: 10.1111/jmp.12510] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/30/2020] [Accepted: 12/30/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND Biomedical research has recently focused on developing new models of human disease by implementing genome-editing strategies in non-human primates (NHPs) to introduce relevant gene mutations. There is a need to establish objective semen evaluation methods to select sires for in vitro fertilization to perform germline editing in embryos. METHODS Sperm motility kinematic parameters were evaluated using a computer-assisted semen analysis (CASA) instrument for rhesus macaques (Macaca mulatta), cynomolgus macaques (Macaca fascicularis), and common marmosets (Callithrix jacchus). RESULTS Normative sperm kinematic parameters were established, revealing differences between marmosets and macaques. The impact of season on rhesus macaque sperm motility was modest, where changes in sperm motility related to season were dependent on the individual male. CONCLUSIONS These data provide a baseline of normative kinematic parameters for three captive NHP species, in which implementation of CASA may serve as a tool to evaluate NHP semen quality.
Collapse
Affiliation(s)
| | | | | | - Hayly M. Hinkle
- Wisconsin National Primate Research Center, Madison, WI, USA
| | | | | | | | - Emily L. Sneed
- Wisconsin National Primate Research Center, Madison, WI, USA
| | - Amy W. Moy
- Wisconsin National Primate Research Center, Madison, WI, USA
| | - Thaddeus G. Golos
- Wisconsin National Primate Research Center, Madison, WI, USA
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
8
|
Park JE, Sasaki E. Assisted Reproductive Techniques and Genetic Manipulation in the Common Marmoset. ILAR J 2021; 61:286-303. [PMID: 33693670 PMCID: PMC8918153 DOI: 10.1093/ilar/ilab002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 10/27/2020] [Accepted: 11/05/2020] [Indexed: 12/12/2022] Open
Abstract
Abstract
Genetic modification of nonhuman primate (NHP) zygotes is a useful method for the development of NHP models of human diseases. This review summarizes the recent advances in the development of assisted reproductive and genetic manipulation techniques in NHP, providing the basis for the generation of genetically modified NHP disease models. In this study, we review assisted reproductive techniques, including ovarian stimulation, in vitro maturation of oocytes, in vitro fertilization, embryo culture, embryo transfer, and intracytoplasmic sperm injection protocols in marmosets. Furthermore, we review genetic manipulation techniques, including transgenic strategies, target gene knock-out and knock-in using gene editing protocols, and newly developed gene-editing approaches that may potentially impact the production of genetically manipulated NHP models. We further discuss the progress of assisted reproductive and genetic manipulation techniques in NHP; future prospects on genetically modified NHP models for biomedical research are also highlighted.
Collapse
Affiliation(s)
- Jung Eun Park
- Department of Neurobiology, University of Pittsburgh, School of Medicine in Pittsburgh, Pennsylvania, USA
| | - Erika Sasaki
- Department of Marmoset Biology and Medicine, Central Institute for Experimental Animals in Kawasaki, Kanagawa, Japan
| |
Collapse
|
9
|
Strelchenko NS, Schmidt JK, Mean KD, Schotzko ML, Golos TG, Slukvin II. Cryopreservation of Mauritian Cynomolgus Macaque ( Macaca fascicularis) Sperm in Chemically Defined Medium. JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE 2020; 59:681-686. [PMID: 32878681 DOI: 10.30802/aalas-jaalas-20-000059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The objective of this study was to optimize cryopreservation of sperm from Mauritian cynomolgus macaques (MCM) in defined conditions. Sperm viability and motility were compared between sperm cryopreserved in chemically-defined freezing media with variable osmolarity and the presence of either ethylene glycol or glycerol. The highest percentage viability (after freeze-thaw) was seen in sperm samples that were cryopreserved in medium with an osmolarity of 310 mOsm, while higher osmolarities markedly decreased sperm viability. Ethylene glycol and glycerol at concentrations of 4.6% and 5%, respectively, preserved sperm viability to an equivalent degree. Although higher motility rates and higher straight-line velocities were observed in sperm samples frozen in glycerol compared with ethylene glycol, these differences were not statistically significant. Thawed sperm frozen in defined conditions with glycerol were capable of fertilizing MCM oocytes in vitro, with development to the blastocyst stage. The protocol described here provides an effective method for cryopreservation of sperm to facilitate subsequent in vitro fertilization and genome editing of embryos in MCM species.
Collapse
Affiliation(s)
- Nick S Strelchenko
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - Jenna Kropp Schmidt
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - Katherine D Mean
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - Michele L Schotzko
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - Thaddeus G Golos
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin; Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin; Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Igor I Slukvin
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin; Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin; Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin;,
| |
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: 2.3] [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
|
Chansel‐Debordeaux L, Bezard E. Local transgene expression and whole-body transgenesis to model brain diseases in nonhuman primate. Animal Model Exp Med 2019; 2:9-17. [PMID: 31016282 PMCID: PMC6431118 DOI: 10.1002/ame2.12055] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 12/10/2018] [Indexed: 12/26/2022] Open
Abstract
Animal model is an essential tool in the life sciences research, notably in understanding the pathogenesis of the diseases and for further therapeutic intervention success. Rodents have been the most frequently used animals to model human disease since the establishment of gene manipulation technique. However, they remain inadequate to fully mimic the pathophysiology of human brain disease, partially due to huge differences between rodents and humans in terms of anatomy, brain function, and social behaviors. Nonhuman primates are more suitable in translational perspective. Thus, genetically modified animals have been generated to investigate neurologic and psychiatric disorders. The classical transgenesis technique is not efficient in that model; so, viral vector-mediated transgene delivery and the new genome-editing technologies have been promoted. In this review, we summarize some of the technical progress in the generation of an ad hoc animal model of brain diseases by gene delivery and real transgenic nonhuman primate.
Collapse
Affiliation(s)
- Lucie Chansel‐Debordeaux
- Institut des Maladies NeurodégénérativesUniversity of BordeauxUMR 5293BordeauxFrance
- CNRSInstitut des Maladies NeurodégénérativesUMR 5293BordeauxFrance
- CHU BordeauxService de Biologie de la reproduction‐CECOSBordeauxFrance
| | - Erwan Bezard
- Institut des Maladies NeurodégénérativesUniversity of BordeauxUMR 5293BordeauxFrance
- CNRSInstitut des Maladies NeurodégénérativesUMR 5293BordeauxFrance
| |
Collapse
|
12
|
Kropp J, Di Marzo A, Golos T. Assisted reproductive technologies in the common marmoset: an integral species for developing nonhuman primate models of human diseases. Biol Reprod 2018; 96:277-287. [PMID: 28203717 DOI: 10.1095/biolreprod.116.146514] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 01/03/2017] [Accepted: 01/05/2017] [Indexed: 12/31/2022] Open
Abstract
Generation of nonhuman primate models of human disease conditions will foster the development of novel therapeutic strategies. Callithrix jacchus, or the common marmoset, is a New World, nonhuman primate species that exhibits great reproductive fitness in captivity with an ovarian cycle that can be easily managed with pharmacological agents. This characteristic, among others, provides an opportunity to employ assisted reproductive technologies to generate embryos that can be genetically manipulated to create a variety of nonhuman primate models for human disease. Here, we review methods to synchronize the marmoset ovarian cycle and stimulate oocyte donors, and compare various protocols for in vitro production of embryos. In light of advances in genomic editing, recent approaches used to generate transgenic or genetically edited embryos in the marmoset and also future perspective are reviewed.
Collapse
Affiliation(s)
- Jenna Kropp
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Andrea Di Marzo
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Thaddeus Golos
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
13
|
Deficiency of PRKD2 triggers hyperinsulinemia and metabolic disorders. Nat Commun 2018; 9:2015. [PMID: 29789568 PMCID: PMC5964083 DOI: 10.1038/s41467-018-04352-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 04/23/2018] [Indexed: 01/21/2023] Open
Abstract
Hyperinsulinemia is the earliest symptom of insulin resistance (IR), but a causal relationship between the two remains to be established. Here we show that a protein kinase D2 (PRKD2) nonsense mutation (K410X) in two rhesus monkeys with extreme hyperinsulinemia along with IR and metabolic defects by using extreme phenotype sampling and deep sequencing analyses. This mutation reduces PRKD2 at both the mRNA and the protein levels. Taking advantage of a PRKD2-KO mouse model, we demonstrate that PRKD2 deletion triggers hyperinsulinemia which precedes to IR and metabolic disorders in the PRKD2 ablation mice. PRKD2 deficiency promotes β-cell insulin secretion by increasing the expression and activity of L-type Ca2+ channels and subsequently augmenting high glucose- and membrane depolarization-induced Ca2+ influx. Altogether, these results indicate that down-regulation of PRKD2 is involved in the pathogenesis of hyperinsulinemia which, in turn, results in IR and metabolic disorders. Hyperinsulinemia can precede the development of insulin resistance. Here the authors identify a PKD2 mutation that leads to hyperinsulinemia and insulin resistance in Rhesus monkey and show that PKD2 deficiency promotes beta cell insulin secretion by activating L-type Ca2+ channels.
Collapse
|
14
|
Chang TA, Bondarenko GI, Gerami-Naini B, Drenzek JG, Durning M, Garthwaite MA, Schmidt JK, Golos TG. Trophoblast differentiation, invasion and hormone secretion in a three-dimensional in vitro implantation model with rhesus monkey embryos. Reprod Biol Endocrinol 2018; 16:24. [PMID: 29548332 PMCID: PMC5857108 DOI: 10.1186/s12958-018-0340-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 03/04/2018] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND The initiation of primate embryo invasion into the endometrium and the formation of the placenta from trophoblasts, fetal mesenchyme, and vascular components are essential for the establishment of a successful pregnancy. The mechanisms which direct morphogenesis of the chorionic villi, and the interactions between trophectoderm-derived trophoblasts and the fetal mesenchyme to direct these processes during placentation are not well understood due to a dearth of systems to examine and manipulate real-time primate implantation. Here we describe an in vitro three-dimensional (3-D) model to study implantation which utilized IVF-generated rhesus monkey embryos cultured in a Matrigel explant system. METHODS Blastocyst stage embryos were embedded in a 3-D microenvironment of a Matrigel carrier and co-cultured with a feeder layer of cells generating conditioned medium. Throughout the course of embryo co-culture embryo growth and secretions were monitored. Embedded embryos were then sectioned and stained for markers of trophoblast function and differentiation. RESULTS Signs of implantation were observed including enlargement of the embryo mass, and invasion and proliferation of trophoblast outgrowths. Expression of chorionic gonadotropin defined by immunohistochemical staining, and secretion of chorionic gonadotropin and progesterone coincident with the appearance of trophoblast outgrowths, supported the conclusion that a trophoblast cell lineage formed from implanted embryos. Positive staining for selected markers including Ki67, MHC class I, NeuN, CD31, vonWillebrand Factor and Vimentin, suggest growth and differentiation of the embryo following embedding. CONCLUSIONS This 3-D in vitro system will facilitate further study of primate embryo biology, with potential to provide a platform for study of genes related to implantation defects and trophoblast differentiation.
Collapse
Affiliation(s)
- T Arthur Chang
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Court, Madison, Wisconsin, 53715-1299, USA
- Present address: Department of Obstetrics and Gynecology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Gennadiy I Bondarenko
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Court, Madison, Wisconsin, 53715-1299, USA
- Present address: Covance Laboratories, Madison, WI, USA
| | - Behzad Gerami-Naini
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Court, Madison, Wisconsin, 53715-1299, USA
- Present address: School of Dental Medicine, Tufts University, Boston, MA, USA
| | - Jessica G Drenzek
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Court, Madison, Wisconsin, 53715-1299, USA
- Present address: Illumina-Madison, Madison, WI, USA
| | - Maureen Durning
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Court, Madison, Wisconsin, 53715-1299, USA
| | - Mark A Garthwaite
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Court, Madison, Wisconsin, 53715-1299, USA
| | - Jenna Kropp Schmidt
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Court, Madison, Wisconsin, 53715-1299, USA
| | - Thaddeus G Golos
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Court, Madison, Wisconsin, 53715-1299, USA.
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Obstetrics and Gynecology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
15
|
Genetic engineering in nonhuman primates for human disease modeling. J Hum Genet 2017; 63:125-131. [PMID: 29203824 PMCID: PMC8075926 DOI: 10.1038/s10038-017-0351-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 09/01/2017] [Accepted: 09/06/2017] [Indexed: 01/29/2023]
Abstract
Nonhuman primate (NHP) experimental models have contributed greatly to human health research by assessing the safety and efficacy of newly developed drugs, due to their physiological and anatomical similarities to humans. To generate NHP disease models, drug-inducible methods, and surgical treatment methods have been employed. Recent developments in genetic and developmental engineering in NHPs offer new options for producing genetically modified disease models. Moreover, in recent years, genome-editing technology has emerged to further promote this trend and the generation of disease model NHPs has entered a new era. In this review, we summarize the generation of conventional disease model NHPs and discuss new solutions to the problem of mosaicism in genome-editing technology.
Collapse
|
16
|
Niu Y, Li T, Ji W. Paving the road for biomedicine: genome editing and stem cells in primates. Natl Sci Rev 2017. [DOI: 10.1093/nsr/nwx094] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Yuyu Niu
- Yunnan Key Laboratory of Primate Biomedicine Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, China
| | - Tianqing Li
- Yunnan Key Laboratory of Primate Biomedicine Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, China
| | - Weizhi Ji
- Yunnan Key Laboratory of Primate Biomedicine Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, China
| |
Collapse
|
17
|
Skoda G, Hoffmann OI, Gócza E, Bodrogi L, Kerekes A, Bösze Z, Hiripi L. Placenta-specific gene manipulation in rabbits. J Biotechnol 2017; 259:86-90. [PMID: 28778693 DOI: 10.1016/j.jbiotec.2017.07.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 07/30/2017] [Accepted: 07/30/2017] [Indexed: 10/19/2022]
Abstract
Lentiviral gene constructs can be efficiently and specifically delivered to trophoblast cell lineages in rodents. In vivo genetic manipulation of trophoblast cell lines enables functional and developmental studies in the placenta. In this report we show that genetic modification can be produced in the extraembryonic tissues of rabbits by lentiviral gene constructs. When 8-16 cell stage embryos were injected with lentiviral particles, strong reporter gene expression resulted in the rabbit placenta. The expression pattern displayed some mosaicism. A strikingly high degree of mosaic GFP expression was detected in some parts of the yolk sac, which is a hypoblast-derived tissue. Whereas expression of the reporter gene construct was detected in placentas and yolk sacs, fetuses never expressed the transgene. As rabbits are an ideal model for functional studies in the placenta, our method would open new possibilities in rabbit biotechnology and placentation studies.
Collapse
Affiliation(s)
- Gabriella Skoda
- Department of Animal Biotechnology, NAIC-Agricultural Biotechnology Institute, Gödöllö, Hungary
| | - Orsolya Ivett Hoffmann
- Department of Animal Biotechnology, NAIC-Agricultural Biotechnology Institute, Gödöllö, Hungary
| | - Elen Gócza
- Department of Animal Biotechnology, NAIC-Agricultural Biotechnology Institute, Gödöllö, Hungary
| | - Lilla Bodrogi
- Department of Animal Biotechnology, NAIC-Agricultural Biotechnology Institute, Gödöllö, Hungary
| | - Andrea Kerekes
- Department of Animal Biotechnology, NAIC-Agricultural Biotechnology Institute, Gödöllö, Hungary
| | - Zsuzsanna Bösze
- Department of Animal Biotechnology, NAIC-Agricultural Biotechnology Institute, Gödöllö, Hungary
| | - Laszlo Hiripi
- Department of Animal Biotechnology, NAIC-Agricultural Biotechnology Institute, Gödöllö, Hungary.
| |
Collapse
|
18
|
Izpisua Belmonte JC, Callaway EM, Caddick SJ, Churchland P, Feng G, Homanics GE, Lee KF, Leopold DA, Miller CT, Mitchell JF, Mitalipov S, Moutri AR, Movshon JA, Okano H, Reynolds JH, Ringach D, Sejnowski TJ, Silva AC, Strick PL, Wu J, Zhang F. Brains, genes, and primates. Neuron 2015; 86:617-31. [PMID: 25950631 DOI: 10.1016/j.neuron.2015.03.021] [Citation(s) in RCA: 193] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
One of the great strengths of the mouse model is the wide array of genetic tools that have been developed. Striking examples include methods for directed modification of the genome, and for regulated expression or inactivation of genes. Within neuroscience, it is now routine to express reporter genes, neuronal activity indicators, and opsins in specific neuronal types in the mouse. However, there are considerable anatomical, physiological, cognitive, and behavioral differences between the mouse and the human that, in some areas of inquiry, limit the degree to which insights derived from the mouse can be applied to understanding human neurobiology. Several recent advances have now brought into reach the goal of applying these tools to understanding the primate brain. Here we describe these advances, consider their potential to advance our understanding of the human brain and brain disorders, discuss bioethical considerations, and describe what will be needed to move forward.
Collapse
Affiliation(s)
- Juan Carlos Izpisua Belmonte
- Gene Expression Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Edward M Callaway
- Systems Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Sarah J Caddick
- The Gatsby Charitable Foundation, The Peak, 5 Wilton Road, London SW1V 1AP, UK
| | - Patricia Churchland
- Department of Philosophy, University of California, San Diego, 1500 Gilman Drive, La Jolla, CA 92093, USA
| | - Guoping Feng
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, 43 Vassar Street, Cambridge, MA 02139, USA
| | - Gregg E Homanics
- Department of Anesthesiology and Pharmacology and Department of Chemical Biology, University of Pittsburgh, 6060 Biomedical Science Tower 3, Pittsburgh, PA 15261, USA
| | - Kuo-Fen Lee
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - David A Leopold
- Section on Cognitive Neurophysiology and Imaging, Laboratory of Neuropsychology, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20192, USA
| | - Cory T Miller
- Department of Psychology and Neurosciences Graduate Program, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Jude F Mitchell
- Brain and Cognitive Sciences, Meliora Hall, Box 270268, University of Rochester, Rochester, NY 14627-0268, USA
| | - Shoukhrat Mitalipov
- Center for Embryonic Cell and Gene Therapy, Oregon Health and Science University, 3303 S.W. Bond Avenue, Portland, OR 97239, USA; Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health and Science University, 505 N.W. 185th Avenue, Beaverton, OR 97006, USA
| | - Alysson R Moutri
- School of Medicine, Department of Pediatrics/Rady Children's Hospital San Diego, and Department of Cellular and Molecular Medicine, Stem Cell Program, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - J Anthony Movshon
- Center for Neural Science, New York University, New York, NY 10003, USA
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; Laboratory for Marmoset Neural Architecture, Brain Science Institute RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - John H Reynolds
- Systems Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | - Dario Ringach
- Department of Neurobiology and Department of Psychology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 92093, USA
| | - Terrence J Sejnowski
- Computational Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Afonso C Silva
- Laboratory of Functional and Molecular Imaging, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 49 Convent Drive, MSC 1065, Building 49, Room 3A72, Bethesda, MD 20892-1065, USA
| | - Peter L Strick
- Brain Institute and Center for the Neural Basis of Cognition, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Research Service, Department of Veterans Affairs Medical Center, Pittsburgh, PA 15261, USA
| | - Jun Wu
- Gene Expression Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Feng Zhang
- Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA 02142, USA; McGovern Institute for Brain Research at MIT, 43 Vassar Street, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, 7 Massachusetts Avenue, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, 7 Massachusetts Avenue, Cambridge, MA 02139, USA; Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, 415 Main Street, Cambridge, MA 02142, USA
| |
Collapse
|
19
|
Sasaki E. Prospects for genetically modified non-human primate models, including the common marmoset. Neurosci Res 2015; 93:110-5. [PMID: 25683291 DOI: 10.1016/j.neures.2015.01.011] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 10/03/2014] [Accepted: 10/07/2014] [Indexed: 01/01/2023]
Abstract
Genetically modified mice have contributed much to studies in the life sciences. In some research fields, however, mouse models are insufficient for analyzing the molecular mechanisms of pathology or as disease models. Often, genetically modified non-human primate (NHP) models are desired, as they are more similar to human physiology, morphology, and anatomy. Recent progress in studies of the reproductive biology in NHPs has enabled the introduction of exogenous genes into NHP genomes or the alteration of endogenous NHP genes. This review summarizes recent progress in the production of genetically modified NHPs, including the common marmoset, and future perspectives for realizing genetically modified NHP models for use in life sciences research.
Collapse
Affiliation(s)
- Erika Sasaki
- Advanced Research Center, Keio University, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan; Center of Applied Developmental Biology, Central Institute for Experimental Animals, 3-25-12 Tonomachi, Kawasaki, Kanagawa 210-0821, Japan.
| |
Collapse
|
20
|
TALEN-mediated gene mutagenesis in rhesus and cynomolgus monkeys. Cell Stem Cell 2014; 14:323-328. [PMID: 24529597 DOI: 10.1016/j.stem.2014.01.018] [Citation(s) in RCA: 163] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 01/21/2014] [Accepted: 01/31/2014] [Indexed: 12/16/2022]
Abstract
Recent advances in gene editing technology have introduced the potential for application of mutagenesis approaches in nonhuman primates to model human development and disease. Here we report successful TALEN-mediated mutagenesis of an X-linked, Rett syndrome (RTT) gene, methyl-CpG binding protein 2 (MECP2), in both rhesus and cynomolgus monkeys. Microinjection of MECP2-targeting TALEN plasmids into rhesus and cynomolgus zygotes leads to effective gene editing of MECP2 with no detected off-target mutagenesis. Male rhesus (2) and cynomolgous (1) fetuses carrying MECP2 mutations in various tissues including testes were miscarried during midgestation, consistent with RTT-linked male embryonic lethality in humans. One live delivery of a female cynomolgus monkey occurred after 162 days of gestation, with abundant MECP2 mutations in peripheral tissues. We conclude that TALEN-mediated mutagenesis can be an effective tool for genetic modeling of human disease in nonhuman primates.
Collapse
|
21
|
Liu C, Wang L, Li W, Zhang X, Tian Y, Zhang N, He S, Chen T, Huang J, Liu M. Highly efficient generation of transgenic sheep by lentivirus accompanying the alteration of methylation status. PLoS One 2013; 8:e54614. [PMID: 23382924 PMCID: PMC3558511 DOI: 10.1371/journal.pone.0054614] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 12/13/2012] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Low efficiency of gene transfer and silence of transgene expression are the critical factors hampering the development of transgenic livestock. Recently, transfer of recombinant lentivirus has been demonstrated to be an efficient transgene delivery method in various animals. However, the lentiviral transgenesis and the methylation status of transgene in sheep have not been well addressed. METHODOLOGY/PRINCIPLE FINDINGS EGFP transgenic sheep were generated by injecting recombinant lentivirus into zygotes. Of the 13 lambs born, 8 carried the EGFP transgene, and its chromosomal integration was identified in all tested tissues. Western blotting showed that GFP was expressed in all transgenic founders and their various tissues. Analysis of CpG methylation status of CMV promoter by bisulfate sequencing unraveled remarkable variation of methylation levels in transgenic sheep. The average methylation levels ranged from 37.6% to 79.1% in the transgenic individuals and 34.7% to 83% in the tested tissues. Correlative analysis of methylation status with GFP expression revealed that the GFP expression level was inversely correlated with methylation density. The similar phenomenon was also observed in tested tissues. Transgene integration determined by Southern blotting presented multiple integrants ranging from 2 to 6 copies in the genome of transgenic sheep. CONCLUSIONS/SIGNIFICANCE Injection of lentiviral transgene into zygotes could be a promising efficient gene delivery system to generate transgenic sheep and achieved widespread transgene expression. The promoter of integrants transferred by lentiviral vector was subjected to dramatic alteration of methylation status and the transgene expression level was inversely correlative with promoter methylation density. Our work illustrated for the first time that generation of transgenic sheep by injecting recombinant lentivirus into zygote could be an efficient tool to improve sheep performance by genetic modification.
Collapse
Affiliation(s)
- Chenxi Liu
- Xinjiang Laboratory of Animal Biotechnology, Urumqi, Xinjiang, China
- Key Laboratory of Genetics, Breeding and Reproduction of Grass Feeding Livestock, Ministry of Agriculture, Urumqi, Xinjiang, China
- College of Life Science and Technology, Xinjiang University, Urumqi, Xinjiang, China
| | - Liqin Wang
- Xinjiang Laboratory of Animal Biotechnology, Urumqi, Xinjiang, China
- Key Laboratory of Genetics, Breeding and Reproduction of Grass Feeding Livestock, Ministry of Agriculture, Urumqi, Xinjiang, China
- Animal Biotechnology Research Center, Xinjiang Academy of Animal Science, Urumqi, Xinjiang, China
| | - Wenrong Li
- Xinjiang Laboratory of Animal Biotechnology, Urumqi, Xinjiang, China
- Key Laboratory of Genetics, Breeding and Reproduction of Grass Feeding Livestock, Ministry of Agriculture, Urumqi, Xinjiang, China
- Animal Biotechnology Research Center, Xinjiang Academy of Animal Science, Urumqi, Xinjiang, China
| | - Xuemei Zhang
- Xinjiang Laboratory of Animal Biotechnology, Urumqi, Xinjiang, China
- Key Laboratory of Genetics, Breeding and Reproduction of Grass Feeding Livestock, Ministry of Agriculture, Urumqi, Xinjiang, China
- Animal Biotechnology Research Center, Xinjiang Academy of Animal Science, Urumqi, Xinjiang, China
| | - Yongzhi Tian
- Xinjiang Laboratory of Animal Biotechnology, Urumqi, Xinjiang, China
- Key Laboratory of Genetics, Breeding and Reproduction of Grass Feeding Livestock, Ministry of Agriculture, Urumqi, Xinjiang, China
- Animal Biotechnology Research Center, Xinjiang Academy of Animal Science, Urumqi, Xinjiang, China
| | - Ning Zhang
- Xinjiang Laboratory of Animal Biotechnology, Urumqi, Xinjiang, China
- Key Laboratory of Genetics, Breeding and Reproduction of Grass Feeding Livestock, Ministry of Agriculture, Urumqi, Xinjiang, China
- Animal Biotechnology Research Center, Xinjiang Academy of Animal Science, Urumqi, Xinjiang, China
| | - Sangang He
- Xinjiang Laboratory of Animal Biotechnology, Urumqi, Xinjiang, China
- Key Laboratory of Genetics, Breeding and Reproduction of Grass Feeding Livestock, Ministry of Agriculture, Urumqi, Xinjiang, China
- Animal Biotechnology Research Center, Xinjiang Academy of Animal Science, Urumqi, Xinjiang, China
| | - Tong Chen
- Xinjiang Laboratory of Animal Biotechnology, Urumqi, Xinjiang, China
- Key Laboratory of Genetics, Breeding and Reproduction of Grass Feeding Livestock, Ministry of Agriculture, Urumqi, Xinjiang, China
- Animal Biotechnology Research Center, Xinjiang Academy of Animal Science, Urumqi, Xinjiang, China
| | - Juncheng Huang
- Xinjiang Laboratory of Animal Biotechnology, Urumqi, Xinjiang, China
- Key Laboratory of Genetics, Breeding and Reproduction of Grass Feeding Livestock, Ministry of Agriculture, Urumqi, Xinjiang, China
- Animal Biotechnology Research Center, Xinjiang Academy of Animal Science, Urumqi, Xinjiang, China
| | - Mingjun Liu
- Xinjiang Laboratory of Animal Biotechnology, Urumqi, Xinjiang, China
- Key Laboratory of Genetics, Breeding and Reproduction of Grass Feeding Livestock, Ministry of Agriculture, Urumqi, Xinjiang, China
- Animal Biotechnology Research Center, Xinjiang Academy of Animal Science, Urumqi, Xinjiang, China
- * E-mail:
| |
Collapse
|
22
|
Chen Y, Niu Y, Ji W. Transgenic nonhuman primate models for human diseases: approaches and contributing factors. J Genet Genomics 2012; 39:247-51. [PMID: 22749011 DOI: 10.1016/j.jgg.2012.04.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Revised: 04/29/2012] [Accepted: 04/30/2012] [Indexed: 11/25/2022]
Abstract
Nonhuman primates (NHPs) provide powerful experimental models to study human development, cognitive functions and disturbances as well as complex behavior, because of their genetic and physiological similarities to humans. Therefore, NHPs are appropriate models for the study of human diseases, such as neurodegenerative diseases including Parkinson's, Alzheimer's and Huntington's diseases, which occur as a result of genetic mutations. However, such diseases afflicting humans do not occur naturally in NHPs. So transgenic NHPs need to be established to understand the etiology of disease pathology and pathogenesis. Compared to rodent genetic models, the generation of transgenic NHPs for human diseases is inefficient, and only a transgenic monkey model for Huntington's disease has been reported. This review focuses on potential approaches and contributing factors for generating transgenic NHPs to study human diseases.
Collapse
Affiliation(s)
- Yongchang Chen
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming 650500, China
| | | | | |
Collapse
|
23
|
CHEN YONGCHANG, NIU YUYU, YANG SHIHUA, HE XIECHAO, JI SHAOHUI, SI WEI, TANG XIANGHUI, XIE YUNHUA, WANG HONG, LU YONGQING, ZHOU QI, JI WEIZHI. The Available Time Window for Embryo Transfer in the Rhesus Monkey (Macaca mulatta). Am J Primatol 2012; 74:165-73. [DOI: 10.1002/ajp.21017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - QI ZHOU
- State Key Laboratory of Reproductive Biology; Institute of Zoology; Chinese Academy of Sciences; Beijing; China
| | | |
Collapse
|
24
|
Katayama K, Furuki R, Yokoyama H, Kaneko M, Tachibana M, Yoshida I, Nagase H, Tanaka K, Sakurai F, Mizuguchi H, Nakagawa S, Nakanishi T. Enhanced in vivo gene transfer into the placenta using RGD fiber-mutant adenovirus vector. Biomaterials 2011; 32:4185-93. [PMID: 21411139 DOI: 10.1016/j.biomaterials.2011.02.038] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Accepted: 02/19/2011] [Indexed: 11/19/2022]
Abstract
Among viral vectors, the fiber-mutant adenovirus vector carrying the Arg-Gly-Asp (RGD) peptide sequence (Ad-RGD) seems to have potential for both clinical gene therapy and basic research. As a part of a thorough evaluation of Ad-RGD in preclinical studies, we designed an experiment to investigate in detail the distribution of Ad-RGD compared with conventional adenovirus vector (WT-Ad) in pregnant mice. Surprisingly, Ad-RGD had substantial placental tropism, at 10-100 times that of WT-Ad. Transgene expression was sustained for at least 7 days, and Ad-RGD expressing firefly luciferase or red fluorescent protein has so far caused no placental dysfunction leading to fetal death. Ad-RGD showed high levels of transduction efficiency in in vitro-differentiated trophoblast stem cells, in which higher expression of αvβ3 integrin than in undifferentiated cells was observed. Our results suggest that the use of Ad-RGD or another RGD-mediated targeting strategy holds promise for drug delivery to the placenta.
Collapse
Affiliation(s)
- Kazufumi Katayama
- Department of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Renaud SJ, Karim Rumi MA, Soares MJ. Review: Genetic manipulation of the rodent placenta. Placenta 2011; 32 Suppl 2:S130-5. [PMID: 21256588 DOI: 10.1016/j.placenta.2010.12.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Revised: 12/10/2010] [Accepted: 12/13/2010] [Indexed: 10/18/2022]
Abstract
The principal role of the placenta is the maintenance of pregnancy and promotion of fetal growth and viability. The use of transgenic rodents has greatly enhanced our understanding of placental development and function. However, embryonic lethality is often a confounding variable in determining whether a genetic modification adversely affected placental development. In these cases, it is beneficial to specifically manipulate the placental genome. The purpose of this review is to summarize available methodologies for specific genetic modification of the rodent placenta. By restricting genetic alterations to the trophoblast lineage, it is possible to gain a deeper understanding of placental development that perhaps will lead to gene-targeted therapies to rescue irregular placentation in transgenic animals or in women at high-risk for placenta-associated pregnancy complications.
Collapse
Affiliation(s)
- S J Renaud
- Institute for Reproductive Health and Regenerative Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | | | | |
Collapse
|
26
|
Transgenic rabbit production with simian immunodeficiency virus-derived lentiviral vector. Transgenic Res 2010; 19:799-808. [PMID: 20069454 DOI: 10.1007/s11248-009-9356-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Accepted: 12/23/2009] [Indexed: 10/20/2022]
Abstract
Transgenic rabbit is the preferred disease model of atherosclerosis, lipoprotein metabolism and cardiovascular diseases since upon introducing genetic mutations of human genes, rabbit models reflect human physiological and pathological states more accurately than mouse models. Beyond that, transgenic rabbits are also used as bioreactors to produce pharmaceutical proteins in their milk. Since in the laboratory rabbit the conventional transgenesis has worked with the same low efficiency in the last twenty five years and truly pluripotent embryonic stem cells are not available to perform targeted mutagenesis, our aim was to adapt lentiviral transgenesis to this species. A simian immunodeficiency virus based replication defective lentiviral vector was used to create transgenic rabbit through perivitelline space injection of fertilized oocytes. The enhanced green fluorescent protein (GFP) gene was placed under the ubiquitous CAG promoter. Transgenic founder rabbits showed mosaic pattern of GFP expression. Transgene integration and expression was revealed in tissues derived from all three primary germ layers. Transgene expression was detected in the developing sperm cells and could get through the germ line without epigenetic silencing, albeit with very low frequency. Our data show for the first time, that lentiviral transgenesis could be a feasible and viable alternative method to create genetically modified laboratory rabbit.
Collapse
|
27
|
Chan AWS. Transgenic primate research paves the path to a better animal model: are we a step closer to curing inherited human genetic disorders? J Mol Cell Biol 2009; 1:13-4. [PMID: 19671628 DOI: 10.1093/jmcb/mjp009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
While the advancement of transgenic primate models has led to a new era in modeling human conditions and has a clear impact on elucidating the mechanism of human genetic diseases, some thoughts should be considered if non-human primates are the appropriate model.
Collapse
Affiliation(s)
- Anthony W S Chan
- Yerkes National Primate Research Center, Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30329, USA.
| |
Collapse
|
28
|
Generation of transgenic non-human primates with germline transmission. Nature 2009; 459:523-7. [PMID: 19478777 DOI: 10.1038/nature08090] [Citation(s) in RCA: 524] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2008] [Accepted: 04/30/2009] [Indexed: 11/08/2022]
Abstract
The common marmoset (Callithrix jacchus) is increasingly attractive for use as a non-human primate animal model in biomedical research. It has a relatively high reproduction rate for a primate, making it potentially suitable for transgenic modification. Although several attempts have been made to produce non-human transgenic primates, transgene expression in the somatic tissues of live infants has not been demonstrated by objective analyses such as polymerase chain reaction with reverse transcription or western blots. Here we show that the injection of a self-inactivating lentiviral vector in sucrose solution into marmoset embryos results in transgenic common marmosets that expressed the transgene in several organs. Notably, we achieved germline transmission of the transgene, and the transgenic offspring developed normally. The successful creation of transgenic marmosets provides a new animal model for human disease that has the great advantage of a close genetic relationship with humans. This model will be valuable to many fields of biomedical research.
Collapse
|
29
|
Abstract
Genetically engineered monkeys carrying a foreign gene that is passed on to their offspring provide a potentially valuable bridge between mouse models of disease and treatment for human disorders.
Collapse
|
30
|
Gómez MC, Pope CE, Kutner RH, Ricks DM, Lyons LA, Ruhe MT, Dumas C, Lyons J, Dresser BL, Reiser J. Generation of domestic transgenic cloned kittens using lentivirus vectors. CLONING AND STEM CELLS 2009; 11:167-76. [PMID: 19203232 DOI: 10.1089/clo.2008.0054] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The efficient use of somatic cell nuclear transfer (SCNT), in conjunction with genetic modification of donor cells provides a general means to add or inactivate genes in mammals. This strategy has substantially improved the efficacy of producing genetically identical animals carrying mutant genes corresponding to specific human disorders. Lentiviral (LV) vectors have been shown to be well suited for introducing transgenes into cells to be used as donor nuclei for SCNT. In the present study, we established an LV vector-based transgene delivery approach for producing live transgenic domestic cats by SCNT. We have demonstrated that cat fetal fibroblasts can be transduced with EGFP-encoding LV vectors bearing various promoters including the human cytomegalovirus immediate early (hCMV-IE) promoter, the human translation elongation factor 1alpha (hEF-1alpha) promoter and the human ubiquitin C (hUbC) promoter. Among the promoters tested, embryos reconstructed with donor cells transduced with a LV-vector bearing the hUbC promoter displayed sustained transgene expression at the blastocyst stage while embryos reconstructed with LV vector-transduced cells containing hCMV-IE-EGFP or hEF-1alpha-EGFP cassettes did not. After transfer of 291 transgenic cloned embryos into the oviducts of eight recipient domestic cats (mean =36.5 +/- 10.1), three (37.5%) were diagnosed to be pregnant, and a total of six embryos (2.1%) implanted. One live male offspring was delivered by Cesarean section on day 64 of gestation, and two kittens were born dead after premature delivery on day 55. In summary, we report the birth of transgenic cloned kittens produced by LV vector-mediated transduction of donor cells and confirm that cloned kittens express the EGFP reporter transgene in all body tissues.
Collapse
Affiliation(s)
- Martha C Gómez
- Audubon Center for Research of Endangered Species, New Orleans, Louisiana 70131, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Kubisch HM, Gagliardi C, Romero DG, Bunnell BA, Ratterree MS. Kinetics of pronuclear development and the effects of vector type and timing of injection on the efficiency of gene transfer into rhesus macaque embryos. Mol Reprod Dev 2008; 75:1505-14. [PMID: 18361395 DOI: 10.1002/mrd.20901] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
A series of experiments was performed to determine the dynamics of pronuclear development as well as the efficiency of either adenovirus-associated (AAV) or lentivirus-derived vectors to introduce a green fluorescent protein (GFP) reporter gene into rhesus macaque (Macaca mulatta) embryos. Assessment of pronuclear development at various times after fertilization revealed that the appearance of pronuclei was determined by the presence of the first and the timing of the second polar body. The dynamics of pronuclear formation was a significant determinant of whether an oocyte reached the blastocyst stage, however, when the percentage of blastocysts were based on the number of zygotes, the timing of the appearance of polar bodies did not appear to have any effect on subsequent development. Injection of different AAV-derived vectors showed that the serotype of the vector did not affect development or the proportion of transgenic embryos. Moreover, all putative transgenic embryos proved to be expression mosaics. Injection of embryos with lentiviral vectors showed that timing of injection (before or after fertilization) had no effect on subsequent transgene expression, but that the type of reporter gene determined post-injection development and rate of transgenesis. The transfer of embryos following injection of a lentiviral vector into three recipients resulted in one pregnancy which was lost during the second trimester. Analysis of fetal tissues showed ubiquitous presence of the transgene and GFP expression in all tissues examined. These results show that lentivirus-derived vectors can efficiently transform rhesus embryos and are suitable for the generation of transgenic rhesus monkeys.
Collapse
Affiliation(s)
- H M Kubisch
- Tulane National Primate Research Center, Covington, Louisiana 70433, USA.
| | | | | | | | | |
Collapse
|
32
|
Abstract
Lentiviral vectors have become a promising new tool for the establishment of transgenic animals and the manipulation of the mammalian genome. While conventional microinjection-based methods for transgenesis have been successful in generating small and large transgenic animals, their relatively low transgenic efficiency has opened the door for alternative approaches, including lentiviral vectors. Lentiviral vectors are an appealing tool for transgenesis in part because of their ability to incorporate into genomic DNA with high efficiency, especially in cells that are not actively dividing. Lentiviral vector-mediated transgene expression can also be maintained for long periods of time. Recent studies have documented high efficiencies for lentiviral transgenesis, even in animal species and strains, such as NOD/ scid and C57Bl/6 mouse, that are very difficult to manipulate using the standard transgenic techniques. These advantages of the lentiviral vector system have broadened its use as a gene therapy vector to additional applications that include transgenesis and knockdown functional genetics. This review will address the components of the lentiviral vector system and recent successes in lentiviral transgenesis using both male- and female-derived pluripotent cells. The advantages and disadvantages of lentiviral transgenesis vs. other approaches to produce transgenic animals will be compared with regard to efficiency, the ability to promote persistent transgene expression, and the time necessary to generate a sufficient number of animals for phenotyping.
Collapse
Affiliation(s)
- Frank Park
- Department of Medicine, Kidney Disease Center, Medical College of Wisconsin, Wauwatosa, Wisconsin 53226, USA.
| |
Collapse
|
33
|
Malashicheva A, Kanzler B, Tolkunova E, Trono D, Tomilin A. Lentivirus as a tool for lineage-specific gene manipulations. Genesis 2007; 45:456-9. [PMID: 17607693 DOI: 10.1002/dvg.20313] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The trophectoderm (TE) of blastocysts, the first epithelium established in mammalian development, (1) plays signaling, supportive, and patterning functions during preimplantation development, (2) ensures embryo implantation into the uterine wall, and (3) gives rise to extraembryonic tissues essential for embryo patterning and growth after implantation. We show that mouse TE, itself permissive to lentiviral (LV) infection, represents a robust nonpermeable physical barrier to the virus particles, thereby shielding the cells of the inner cell mass from viral infection. This LV feature will allow modulations of gene expression in a lineage-specific manner, thus having significant applications in mouse functional genetics.
Collapse
Affiliation(s)
- Anna Malashicheva
- Department of Developmental Biology, Max-Planck Institute for Immunobiology, Freiburg, Germany
| | | | | | | | | |
Collapse
|
34
|
Georgiades P, Cox B, Gertsenstein M, Chawengsaksophak K, Rossant J. Trophoblast-specific gene manipulation using lentivirus-based vectors. Biotechniques 2007; 42:317-8, 320, 322-5. [PMID: 17390538 DOI: 10.2144/000112341] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The trophoblast layers of the mammalian placenta carry out many complex functions required to pattern the developing embryo and maintain its growth and survival in the uterine environment. Genetic disruption of many gene pathways can result in embryonic lethality because of placental failure, potentially confusing the interpretation of mouse knockout phenotypes. Development of tools to specifically and efficiently manipulate gene expression in the trophoblast lineage would greatly aid understanding of the relative roles of different genetic pathways in the trophoblast versus embryonic lineages. We show that short-term lentivirus-mediated infection of mouse blastocysts can lead to rapid expression of a green fluorescent protein (GFP) transgene specifically in the outer trophoblast progenitors and their later placental derivatives. Efficient trophoblast-specific gene knockdown can also be produced by lentivirus-mediated pol III-driven short hairpin RNA (shRNA) and efficient trophoblast-specific gene knockout by pol II-driven Cre recombinase lentiviral vectors. This lentivirus lineage-specific infection system thus facilitates both gain and loss of function studies during placental development in the mouse and potentially other mammalian species.
Collapse
Affiliation(s)
- Pantelis Georgiades
- Developmental Genetics and Embryology Research Unit, Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus.
| | | | | | | | | |
Collapse
|
35
|
|
36
|
Alves MP, Neuhaus V, Guzylack-Piriou L, Ruggli N, McCullough KC, Summerfield A. Toll-like receptor 7 and MyD88 knockdown by lentivirus-mediated RNA interference to porcine dendritic cell subsets. Gene Ther 2007; 14:836-44. [PMID: 17330086 DOI: 10.1038/sj.gt.3302930] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Sensing of viruses by dendritic cell (DC) pathogen recognition receptors (PRRs) represents a critical event during innate antiviral immune responses. Identification of these PRRs has often posed a problem due to difficulties in performing gene function studies in the naturally targeted hosts. Consequently, we developed a lentivirus (LV)-based strategy for specific gene knockdown in porcine DC. Short hairpin RNAs (shRNAs) were designed, targeting toll-like receptor 7 (TLR7) and the adaptor protein MyD88. As cellular targets, monocyte-derived DC (MoDC) and Flt3 ligand-induced DC (Flt3L-DC), DC precursors including monocytes and haematopoietic stem cells (HSCs) as well as plasmacytoid DCs (pDCs) were employed. Transduction efficiencies ranged from 40 to 95%. The LV-mediated shRNA delivery was functionally active, reducing TLR7 and MyD88 mRNA in MoDC and conventional Flt3L-DC, and blunting the responsiveness to TLR7 ligands in Flt3L-DC. Although infection of MoDC by the LV did neither influence MHC class II and CD80/86 expressions, nor cytokine responses, the infection of Flt3L-DC induced a phenotypic maturation. Furthermore, the interaction of the LV with pDC induced high levels of interferon-alpha. Taken together, these studies characterize the interaction of the LV with different DC subsets and demonstrate the suitability of LV-mediated small interfering RNA delivery for targeting PRR knockout for MoDC and conventional Flt3L-DC.
Collapse
Affiliation(s)
- M P Alves
- Institute of Virology and Immunoprophylaxis, Mittelhäusern, Switzerland
| | | | | | | | | | | |
Collapse
|
37
|
Okada Y, Ueshin Y, Isotani A, Saito-Fujita T, Nakashima H, Kimura K, Mizoguchi A, Oh-Hora M, Mori Y, Ogata M, Oshima RG, Okabe M, Ikawa M. Complementation of placental defects and embryonic lethality by trophoblast-specific lentiviral gene transfer. Nat Biotechnol 2007; 25:233-7. [PMID: 17220877 DOI: 10.1038/nbt1280] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2006] [Accepted: 12/08/2006] [Indexed: 11/09/2022]
Abstract
Placental dysfunction underlies many complications during pregnancy, and better understanding of gene function during placentation could have considerable clinical relevance. However, the lack of a facile method for placenta-specific gene manipulation has hampered investigation of placental organogenesis and the treatment of placental dysfunction. We showed previously that transduction of fertilized mouse eggs with lentiviral vectors leads to transgene expression in both the fetus and the placenta. Here we report placenta-specific gene incorporation by lentiviral transduction of mouse blastocysts after removal of the zona pellucida. All of the placentas analyzed, but none of the fetuses, were transgenic. Application of this method substantially rescued mice deficient in Ets2, Mapk14 (also known as p38alpha) and Mapk1 (also known as Erk2) from embryonic lethality caused by placental defects. Ectopic expression of Mapk11 also complemented Mapk14 deficiency during placentation.
Collapse
Affiliation(s)
- Yuka Okada
- Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Considerable progress has been made in methods for production of transgenic livestock; beginning with pronuclear microinjection over 20 years ago. New methods, including the use of viral vectors, sperm-mediated gene transfer and somatic cell cloning, have overcome many of the limitations of pronuclear microinjection. It is now possible to not only readily make simple insertional genetic modifications, but also to accomplish, more complex, homozygous gene targeting and artificial chromosome transfer in livestock.
Collapse
Affiliation(s)
- J M Robl
- Hematech Inc, Sioux Falls, SD 57106, USA.
| | | | | | | |
Collapse
|
39
|
Vodicka P, Smetana K, Dvoránková B, Emerick T, Xu YZ, Ourednik J, Ourednik V, Motlík J. The Miniature Pig as an Animal Model in Biomedical Research. Ann N Y Acad Sci 2006; 1049:161-71. [PMID: 15965115 DOI: 10.1196/annals.1334.015] [Citation(s) in RCA: 285] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Crucial prerequisites for the development of safe preclinical protocols in biomedical research are suitable animal models that would allow for human-related validation of valuable research information gathered from experimentation with lower mammals. In this sense, the miniature pig, sharing many physiological similarities with humans, offers several breeding and handling advantages (when compared to non-human primates), making it an optimal species for preclinical experimentation. The present review offers several examples taken from current research in the hope of convincing the reader that the porcine animal model has gained massively in importance in biomedical research during the last few years. The adduced examples are taken from the following fields of investigation: (a) the physiology of reproduction, where pig oocytes are being used to study chromosomal abnormalities (aneuploidy) in the adult human oocyte; (b) the generation of suitable organs for xenotransplantation using transgene expression in pig tissues; (c) the skin physiology and the treatment of skin defects using cell therapy-based approaches that take advantage of similarities between pig and human epidermis; and (d) neurotransplantation using porcine neural stem cells grafted into inbred miniature pigs as an alternative model to non-human primates xenografted with human cells.
Collapse
Affiliation(s)
- Petr Vodicka
- Institute of Animal Physiology and Genetics, Department of Physiology of Reproduction, Rumburská 89, Libechov 277 21, Czech Republic.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Contents Transgenic animals are more widely used for various purposes. Applications of animal transgenesis may be divided into three major categories: (i) to obtain information on gene function and regulation as well as on human diseases, (ii) to obtain high value products (recombinant pharmaceutical proteins and xeno-organs for humans) to be used for human therapy, and (iii) to improve animal products for human consumption. All these applications are directly or not related to human health. Animal transgenesis started in 1980. Important improvement of the methods has been made and are still being achieved to reduce cost as well as killing of animals and to improve the relevance of the models. This includes gene transfer and design of reliable vectors for transgene expression. This review describes the state of the art of animal transgenesis from a technical point of view. It also reports some of the applications in the medical field based on the use of transgenic animal models. The advance in the generation of pigs to be used as the source of organs for patients and in the preparation of pharmaceutical proteins from milk and other possible biological fluids from transgenic animals is described. The projects in course aiming at improving animal production by transgenesis are also depicted. Some the specific biosafety and bioethical problems raised by the different applications of transgenesis, including consumption of transgenic animal products are discussed.
Collapse
Affiliation(s)
- L-M Houdebine
- Biologie du Développement et Reproduction, Institut National de la Recherche Agronomique, Jouy-en-Josas Cedex, France.
| |
Collapse
|
41
|
Abstract
First described about a decade ago, lentiviral vectors ('lentivectors') have emerged as potent and versatile tools of gene transfer for basic and applied research and offer exciting perspectives for the field of gene therapy. In the clinic, HIV-based vectors are showing particular promise for delivering therapeutic genes to hematopoietic stem cells (HSCs) and terminally differentiated targets in the central nervous system (CNS). Their flexible design facilitates the accommodation of sophisticated elements of control for the precise tuning of transgene expression. The delivery of small interfering RNAs (siRNAs) and genomic or cDNA libraries and the creation of transgenic animals are the most recent and exciting applications of HIV-based vectors that will help to tackle fundamental issues across wide areas of biology.
Collapse
Affiliation(s)
- Maciej Wiznerowicz
- Frontiers in Genetics, National Center for Competence in Research, School of Life Sciences, Swiss Federal Institute of Technology, Laussane, Switzerland
| | | |
Collapse
|
42
|
Abstract
Transgenic animals are relevant for many fields of modern biomedicine and agriculture. However, the inefficiencies of the presently available techniques--DNA microinjection and retroviral gene transfer--have led to an explosion of costs for transgenics especially in farm animals. The recent success in transferring genes to early embryos of different species (mouse, rat, pig, cattle) by viral vectors derived from lentiviruses, has established lentiviral transgenesis as an exciting alternative to the classical method of DNA microinjection. In addition, lentiviral vectors can be used to transfer genes into embryonic stem cells. Due to its high efficacy and versatility, lentiviral transgenesis should have a big impact on transgenic research.
Collapse
Affiliation(s)
- Alexander Pfeifer
- Department of Pharmacy, Institute for Pharmacology, Center for Drug Research, Ludwig-Maximilians University, Munich, Germany.
| |
Collapse
|
43
|
Affiliation(s)
- Reinhard Fässler
- Max Planck Institute for Biochemistry, Department of Molecular Medicine, Am Klopferspitz 18a, 82152 Martinsried, Germany.
| |
Collapse
|
44
|
Golos TG. Nonhuman primate placental MHC expression: a model for exploring mechanisms of human maternal-fetal immune tolerance. Hum Immunol 2004; 64:1102-9. [PMID: 14602241 DOI: 10.1016/j.humimm.2003.08.349] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Placental contributions to the establishment of maternal-fetal immune tolerance, and placental influences on maturation and vascular development of the endometrium in the human have been difficult to explore directly. Although significant differences exist in organization and relevant gene expression between human and nonprimate placentas, the nonhuman primate has substantial potential to provide insights into the physiology of human pregnancy and maternal-fetal immune tolerance. In this report, we will summarize major histocompatability complex class I gene expression in the nonhuman primate placenta and present progress in characterizing the immune cells resident in the primate endometrium. Finally, we will outline new experimental approaches for modifying placental function now available to move research forward in this field.
Collapse
Affiliation(s)
- Thaddeus G Golos
- Wisconsin National Primate Research Center and Department of Obstetrics and Gynecology, University of Wisconsin Medical School, Madison, WI 53715-1299, USA.
| |
Collapse
|
45
|
Abstract
The assisted reproductive technologies (ARTs) have been used in the production of rhesus monkey offspring at the Oregon National Primate Research Center (ONPRC) and that experience is summarized here. Additionally these technologies serve as a source of oocytes/embryos for monozygotic twinning, embryonic stem (ES) cell derivation and cloning. High fertilization efficiencies were realized with conventional insemination or following the use of intracytoplasmic sperm injection (ICSI) and approximately 50% of the resulting embryos grew in vitro to blastocysts. Both fresh and frozen sperm were employed in fertilization by ICSI and the resulting embryos could be low temperature stored for subsequent thawing and transfer when a synchronized recipient female was available or after shipment to another facility. Following the transfer of up to 3 embryos, an overall pregnancy rate of 30% was achieved with increasing rates dependent upon the number of embryos transferred. Singleton pregnancy outcomes following the transfer of ART produced embryos were similar to those observed in a control group of animals in the timed mated breeding colony at ONPRC. ICSI produced embryos were used in efforts to create monozygotic twins by blastomere separation or blastocyst splitting. While pregnancies were achieved following the transfer of demi-embryos, only one was a twin and it was lost to spontaneous abortion. ICSI produced embryos have also served as the source of blastocysts for the derivation of embryonic stem cells. These pluripotent cells hold potential for cell based therapies and we consider the monkey an important translational model in which to evaluate safety, efficacy and feasibility of regenerative medicine approaches based on the transplantation of stem cell-derived progeny. Finally, efforts to produce genetically-identical monkeys by nuclear transfer have been briefly summarized.
Collapse
Affiliation(s)
- Don P Wolf
- Division of Reproductive Sciences, Oregon National Primate Research Center Beaverton, Oregon, USA.
| |
Collapse
|
46
|
Golos TG. Pregnancy initiation in the rhesus macaque: towards functional manipulation of the maternal-fetal interface. Reprod Biol Endocrinol 2004; 2:35. [PMID: 15200676 PMCID: PMC446212 DOI: 10.1186/1477-7827-2-35] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2004] [Accepted: 06/16/2004] [Indexed: 12/02/2022] Open
Abstract
Nonhuman primates provide an important opportunity to define the mechanisms that contribute to the success of early pregnancy. We have focused for several years now on defining the expression of novel placental major histocompatibility complex (MHC) class I molecules. In parallel, we have used reagents against human immune cell markers to characterize the leukocyte population in the decidua and have demonstrated dynamic changes in these cell populations during the first 5 weeks of gestation. The challenge is to identify the possible role(s) of placental MHC class I in modifying/directing the maternal endometrial or systemic immune system in the post-implantation period. Foremost among the challenges is the difficulty in modifying placental function. In the instance of trophoblast surface proteins, passive immunization studies are feasible, although limitations include the empirical nature of this approach, as well as the inability to modify intracellular function. We have shown that using lentiviral vectors to effect preimplantation gene transfer for transgene expression in the placenta is not only feasible, but of good efficiency. In addition to transgene overexpression, robust approaches for knocking down/knocking out placental gene expression are essential. Recent developments in RNA interference approaches may allow "transient knockout" experiments. While the rhesus monkey has been our model of choice, currently there are limitations in the number of available female rhesus monkeys of reproductive age for research in early pregnancy. It is critical that the technologies for advanced study move forward in other species. The baboon has been used significantly in reproductive tract biology and early pregnancy research and important models have been developed for manipulation of the maternal-fetal interface. Additional characterization of other species, such as the cynomolgus and African green (vervet) monkey is critical. Given the limitations on antigen recognition when using human reagents, we also propose that the development of panels of primate-specific anti-leukocyte antibodies is essential for moving forward nonhuman primate reproductive research.
Collapse
Affiliation(s)
- Thaddeus G Golos
- National Primate Research Center and Department of Obstetrics and Gynecology, University of Wisconsin Medical School, University of Wisconsin-Madison, Madison, WI 53715-1299, USA.
| |
Collapse
|
47
|
Norgren RB. Creation of non-human primate neurogenetic disease models by gene targeting and nuclear transfer. Reprod Biol Endocrinol 2004; 2:40. [PMID: 15200671 PMCID: PMC455690 DOI: 10.1186/1477-7827-2-40] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2004] [Accepted: 06/16/2004] [Indexed: 12/02/2022] Open
Abstract
Genetically modified rhesus macaques are necessary because mouse models are not suitable for a number of important neurogenetic disorders; for example, Kallmann's syndrome, Lesch-Nyhan's disease and Ataxia-Telangiectasia. Mouse models may not be suitable because there may be no mouse ortholog of the human gene of interest, as is the case for Kallmann's syndrome, or because mutant mice do not exhibit the same phenotype observed in humans, as is the the case for Lesch-Nyhan's disease and Ataxia-Telangiectasia. Non-human primate models of neurogenetic diseases are expected to more closely resemble human diseases than existing mouse models. Genetically modified rhesus macaques can be created by modifying the genome of a somatic cell and then transferring the nucleus from this cell to an enucleated oocyte. Random integration of a transgene is sufficient to create models of gain-of-function genetic diseases. Stable expression of green fluorescent protein has been achieved in rhesus macaque fibroblasts. However, gene targeting is necessary to create models of loss-of-function genetic diseases. Several technical challenges must be overcome before null mutant non-human primates can be produced. In our experience, fetal fibroblasts frequently become senescent before selection procedures can be completed. We have overcome this problem by transfecting somatic cells with human telomerase reverse transcriptase. This enzyme extends the telomeres, and lifespan, of somatic cells. Long and accurate polymerase chain reaction can be used to obtain sufficient regions of homology of isogenic rhesus genomic DNA for targeting constructs. This should improve gene targeting efficiency. Gene targeting experiments are currently underway. Null mutant rhesus macaques will likely result in breakthrough advances in the understanding of neurogenetic disease and prove invaluable for preclinical trials of new therapies.
Collapse
Affiliation(s)
- Robert B Norgren
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska, USA.
| |
Collapse
|
48
|
Pfeifer A, Hofmann A, Kessler B, Wolf E. Response to Whitelaw: Lentiviral transgenesis in livestock. Trends Biotechnol 2004. [DOI: 10.1016/j.tibtech.2004.02.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
49
|
Hofmann A, Zakhartchenko V, Weppert M, Sebald H, Wenigerkind H, Brem G, Wolf E, Pfeifer A. Generation of transgenic cattle by lentiviral gene transfer into oocytes. Biol Reprod 2004; 71:405-9. [PMID: 15044266 DOI: 10.1095/biolreprod.104.028472] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The potential benefits of transgenic cattle range from the production of large quantities of pharmaceutically relevant proteins to agricultural improvement. However, the production of transgenic cattle is presently time-consuming and expensive because of the inefficiency of the classical DNA microinjection technique. Here, we report the use of lentiviruses for the efficient generation of transgenic cattle. Initial attempts to produce transgenic cattle by lentiviral infection of preimplantation embryos were not successful. In contrast, infection of bovine oocytes with lentiviral vectors carrying an enhanced green fluorescent protein (eGFP) expression cassette followed by in vitro fertilization resulted in the birth of transgenic calves. Furthermore, all of the calves generated by infection of oocytes were transgenic, and 100% of these animals expressed eGFP as detected by in vivo imaging and Western blotting. In addition, a transgenic calf was produced by infection of fetal fibroblasts followed by nuclear transfer into enucleated oocytes. Taken together, after adjusting lentiviral transgenesis to cattle, unprecedented high transgenesis and expression rates were achieved.
Collapse
Affiliation(s)
- Andreas Hofmann
- Department of Pharmacy, Institute for Pharmacology, Center for Drug Research, Ludwig-Maximilians University, Butenandstrasse 5(C), 81377 Munich, Germany
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Wolf DP, Thormahlen S, Ramsey C, Yeoman RR, Fanton J, Mitalipov S. Use of assisted reproductive technologies in the propagation of rhesus macaque offspring. Biol Reprod 2004; 71:486-93. [PMID: 15044263 DOI: 10.1095/biolreprod.103.025932] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The assisted reproductive technologies (ARTs) as tailored to the production of rhesus monkeys at the Oregon National Primate Research Center (ONPRC) are described. Efficient fertilization of mature oocytes recovered by aspiration from females subjected to follicular stimulation was achieved with fresh or frozen sperm by intracytoplasmic sperm injection (ICSI). Embryo development to the early cleavage stage occurred at high frequency. Cryopreserved embryos showed high postthaw survival and were also transferred in efforts to establish pregnancies. Three methods of transfer were evaluated, two involving embryo placement into the oviduct, laparoscopy and minilaparotomy, and a nonsurgical, transcervical approach that resulted in uterine deposition. Early cleaving embryos (Days 1-4) were transferred into the oviducts of synchronized recipients with optimal results and pregnancy rates of up to 36%. Pregnancy rates were similar when two fresh or frozen embryos were transferred (28- 30%), although more than two embryos had to be thawed to compensate for embryo loss during freeze-thawing. Normal gestational lengths, birth weights, and growth curves were seen with ART-produced infants compared with infants produced by natural mating in the timed mated breeding (TMB) colony at the ONPRC. In 72 singleton pregnancies established following the transfer of ART-produced embryos, the live-birth rate, at 87.5%, was statistically identical to that for the TMB colony. Further development of the ARTs should result in increasing use of these techniques to augment conventional approaches to propagating monkeys, especially those of defined genotypes.
Collapse
Affiliation(s)
- D P Wolf
- Division of Reproductive Sciences, Oregon National Primate Research Center, 505 NW 185th Avenue, Beaverton, OR 97006, USA.
| | | | | | | | | | | |
Collapse
|