1
|
Abe T, Inoue KI, Kiyonari H. Efficient CRISPR/Cas9-mediated knockin of reporter genes in rats at ROSA26 by pronuclear microinjection. Dev Growth Differ 2025. [PMID: 40269535 DOI: 10.1111/dgd.70007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 03/25/2025] [Accepted: 03/30/2025] [Indexed: 04/25/2025]
Abstract
The genetic modification of rats is a key technology for advancing biomedical research on human diseases. CRISPR/Cas9-mediated genome editing enables the generation of knockout rats in a single step, without the need for embryonic stem cells, by directly injecting genome editing components into zygotes. This simplifies the process, reduces costs, and accelerates gene function analysis in rats. However, the insertion of a gene cassette into a target site has remained inefficient, limiting the generation of knockin (KI) rats. To overcome this issue, we developed an optimized method that covers the entire process from zygote harvesting with superovulation to timed microinjection, ensuring the consistent generation of KI rats. We successfully generated four different fluorescent reporter lines at the ROSA26 locus in rats. Our study provides detailed, step-by-step protocols for donor vector design, zygote collection, microinjection, founder screening, and cryopreservation in rats.
Collapse
Affiliation(s)
- Takaya Abe
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Chuou-ku, Kobe, Japan
| | - Ken-Ichi Inoue
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Chuou-ku, Kobe, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Chuou-ku, Kobe, Japan
| |
Collapse
|
2
|
Mizuno-Iijima S, Kawamoto S, Asano M, Mashimo T, Wakana S, Nakamura K, Nishijima KI, Okamoto H, Saito K, Yoshina S, Miwa Y, Nakamura Y, Ohkuma M, Yoshiki A. Mammalian genome research resources available from the National BioResource Project in Japan. Mamm Genome 2024; 35:497-523. [PMID: 39261329 PMCID: PMC11522087 DOI: 10.1007/s00335-024-10063-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 08/08/2024] [Indexed: 09/13/2024]
Abstract
Mammalian genome research has conventionally involved mice and rats as model organisms for humans. Given the recent advances in life science research, to understand complex and higher-order biological phenomena and to elucidate pathologies and develop therapies to promote human health and overcome diseases, it is necessary to utilize not only mice and rats but also other bioresources such as standardized genetic materials and appropriate cell lines in order to gain deeper molecular and cellular insights. The Japanese bioresource infrastructure program called the National BioResource Project (NBRP) systematically collects, preserves, controls the quality, and provides bioresources for use in life science research worldwide. In this review, based on information from a database of papers related to NBRP bioresources, we present the bioresources that have proved useful for mammalian genome research, including mice, rats, other animal resources; DNA-related materials; and human/animal cells and microbes.
Collapse
Affiliation(s)
- Saori Mizuno-Iijima
- Experimental Animal Division, RIKEN BioResource Research Center, Tsukuba, Ibaraki, 305-0074, Japan.
| | - Shoko Kawamoto
- Department of Informatics, National Institute of Genetics, Mishima, Shizuoka, 411-8540, Japan
| | - Masahide Asano
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Tomoji Mashimo
- Division of Animal Genetics, Laboratory Animal Research Center, Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, 108-8639, Japan
| | - Shigeharu Wakana
- Department of Animal Experimentation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Hyogo, 650-0047, Japan
| | - Katsuki Nakamura
- Center for the Evolutionary Origins of Human Behavior, Kyoto University, Inuyama, Aichi, 484-8506, Japan
| | - Ken-Ichi Nishijima
- Avian Bioscience Research Center, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, 464-8601, Japan
| | - Hitoshi Okamoto
- RIKEN Center for Brain Science, Wako, Saitama, 351-0198, Japan
| | - Kuniaki Saito
- Department of Chromosome Science, National Institute of Genetics, Mishima, Shizuoka, 411-8540, Japan
| | - Sawako Yoshina
- Department of Physiology, Tokyo Women's Medical University School of Medicine, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Yoshihiro Miwa
- Gene Engineering Division, RIKEN BioResource Research Center, Tsukuba, Ibaraki, 305-0074, Japan
| | - Yukio Nakamura
- Cell Engineering Division, RIKEN BioResource Research Center, Tsukuba, Ibaraki, 305-0074, Japan
| | - Moriya Ohkuma
- Microbe Division/Japan Collection of Microorganisms, RIKEN BioResource Research Center, Tsukuba, Ibaraki, 305-0074, Japan
| | - Atsushi Yoshiki
- Experimental Animal Division, RIKEN BioResource Research Center, Tsukuba, Ibaraki, 305-0074, Japan.
| |
Collapse
|
3
|
Sato M, Inada E, Saitoh I, Morohoshi K, Nakamura S. Artificial Insemination as a Possible Convenient Tool to Acquire Genome-Edited Mice via In Vivo Fertilization with Engineered Sperm. BIOTECH 2024; 13:45. [PMID: 39584902 PMCID: PMC11587059 DOI: 10.3390/biotech13040045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/04/2024] [Accepted: 11/08/2024] [Indexed: 11/26/2024] Open
Abstract
Advances in genome editing technology have made it possible to create genome-edited (GE) animals, which are useful for identifying isolated genes and producing models of human diseases within a short period of time. The production of GE animals mainly relies on the gene manipulation of pre-implantation embryos, such as fertilized eggs and two-cell embryos, which can usually be achieved by the microinjection of nucleic acids, electroporation in the presence of nucleic acids, or infection with viral vectors, such as adeno-associated viruses. In contrast, GE animals can theoretically be generated by fertilizing ovulated oocytes with GE sperm. However, there are only a few reports showing the successful production of GE animals using GE sperm. Artificial insemination (AI) is an assisted reproduction technology based on the introduction of isolated sperm into the female reproductive tract, such as the uterine horn or oviductal lumen, for the in vivo fertilization of ovulated oocytes. This approach is simpler than the in vitro fertilization-based production of offspring, as the latter always requires an egg transfer to recipient females, which is labor-intensive and time-consuming. In this review, we summarize the various methods for AI reported so far, the history of sperm-mediated gene transfer, a technology to produce genetically engineered animals through in vivo fertilization with sperm carrying exogenous DNA, and finally describe the possibility of AI-mediated creation of GE animals using GE sperm.
Collapse
Affiliation(s)
- Masahiro Sato
- Department of Genome Medicine, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Emi Inada
- Department of Pediatric Dentistry, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Issei Saitoh
- Department of Pediatric Dentistry, Asahi University School of Dentistry, Gifu 501-0296, Japan
| | - Kazunori Morohoshi
- Division of Biomedical Engineering, National Defense Medical College Research Institute, Saitama 359-8513, Japan
| | - Shingo Nakamura
- Division of Biomedical Engineering, National Defense Medical College Research Institute, Saitama 359-8513, Japan
| |
Collapse
|
4
|
Mochida K, Morita K, Sasaoka Y, Morita K, Endo H, Hasegawa A, Asano M, Ogura A. Superovulation with an anti-inhibin monoclonal antibody improves the reproductive performance of rat strains by increasing the pregnancy rate and the litter size. Sci Rep 2024; 14:8294. [PMID: 38670985 PMCID: PMC11052992 DOI: 10.1038/s41598-024-58611-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 04/01/2024] [Indexed: 04/28/2024] Open
Abstract
Rats are multiparous rodents that have been used extensively in research; however, the low reproductive performance of some rat strains hampers the broader use of rats as a biomedical model. In this study, the possibility of increasing the litter size after natural mating in rats through superovulation using an anti-inhibin monoclonal antibody (AIMA) was examined. In outbred Wistar rats, AIMA increased the number of ovulated oocytes by 1.3-fold. AIMA did not affect fertilization and subsequent embryonic development, resulting in a 1.4-fold increase in litter size and a high pregnancy rate (86%). In contrast, conventional superovulation by eCG/hCG administration decreased the pregnancy rate to 6-40% and did not increase the litter size. In inbred Brown Norway rats, AIMA increased the litter size by 1.2-fold, and the pregnancy rate increased more than twice (86% versus 38% in controls). AIMA also increased the litter size by 1.5-fold in inbred Tokai High Avoiders and Fischer 344 rats. AIMA increased the efficiency of offspring production by 1.5-, 2.7-, 1.4-, and 1.4-fold, respectively, in the four rat strains. Thus, AIMA may consistently improve the reproductive performance through natural mating in rats, which could promote the use of AIMA in biomedical research.
Collapse
Affiliation(s)
- Keiji Mochida
- RIKEN BioResource Research Center, Tsukuba, Ibaraki, 305-0074, Japan.
| | - Kohtaro Morita
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Kyoto, 606-8501, Japan
| | - Yoshio Sasaoka
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Kyoto, 606-8501, Japan
| | - Kento Morita
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Kyoto, 606-8501, Japan
| | - Hitoshi Endo
- Center for Molecular Prevention and Environmental Medicine, Tokai University School of Medicine, Isehara, Kanagawa, 259-1193, Japan
| | - Ayumi Hasegawa
- RIKEN BioResource Research Center, Tsukuba, Ibaraki, 305-0074, Japan
| | - Masahide Asano
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Kyoto, 606-8501, Japan.
| | - Atsuo Ogura
- RIKEN BioResource Research Center, Tsukuba, Ibaraki, 305-0074, Japan.
- Graduate School of Life and Environmental Science, University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan.
- RIKEN Cluster for Pioneering Research, Wako, Saitama, 351-0198, Japan.
| |
Collapse
|
5
|
Nakagata N, Nakao S, Mikoda N, Yamaga K, Takeo T. Time elapsed between ovulation and insemination determines the quality of fertilized rat oocytes. J Reprod Dev 2024; 70:123-130. [PMID: 38403585 PMCID: PMC11017092 DOI: 10.1262/jrd.2023-067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 01/29/2024] [Indexed: 02/27/2024] Open
Abstract
Genetically modified rats are valuable models in human disease research. We recently developed an improved system for rat sperm cryopreservation and in vitro fertilization (IVF) that facilitates the efficient production and preservation of genetically modified rats. In the IVF procedure performed using frozen-thawed rat sperm, the IVF schedule is fixed to ensure timely hormone administration and oocyte collection. To enhance the flexibility of the IVF schedule, possible periods of postovulated rat oocytes with normal fertility and developmental abilities should be determined. Therefore, in this study, we examined the fertilization and developmental ability of incubated oocytes 1-13 h after oocyte collection at 9:00 AM. The fertilization rate decreased 7 h after oocyte collection, and abnormally fertilized oocytes appeared 10 h after oocyte collection. The developmental rate also decreased 7 h after oocyte collection; however, live pups were obtained from oocytes 12 h after oocyte collection. In summary, ovulated rat oocytes exhibited a high developmental ability after IVF for up to 4 h after oocyte collection.
Collapse
Affiliation(s)
- Naomi Nakagata
- Division of Reproductive Biotechnology and Innovation, Center for Animal Resources and Development (CARD), Institute of Resource Development and Analysis, Kumamoto University, Kumamoto 860-0811, Japan
| | - Satohiro Nakao
- Division of Reproductive Engineering, Center for Animal Resources and Development (CARD), Institute of Resource Development and Analysis, Kumamoto University, Kumamoto 860-0811, Japan
| | - Nobuyuki Mikoda
- Division of Reproductive Biotechnology and Innovation, Center for Animal Resources and Development (CARD), Institute of Resource Development and Analysis, Kumamoto University, Kumamoto 860-0811, Japan
- Kyudo Co., Ltd., Saga 841-0075, Japan
| | - Katsuma Yamaga
- Division of Reproductive Engineering, Center for Animal Resources and Development (CARD), Institute of Resource Development and Analysis, Kumamoto University, Kumamoto 860-0811, Japan
| | - Toru Takeo
- Division of Reproductive Engineering, Center for Animal Resources and Development (CARD), Institute of Resource Development and Analysis, Kumamoto University, Kumamoto 860-0811, Japan
| |
Collapse
|
6
|
Nakamura K, Seno M, Yoshimura Y, Suzuki O. Long-term culture induces Bax-dependent apoptosis in rat preimplantation embryos. Mol Reprod Dev 2024; 91:e23711. [PMID: 37831754 DOI: 10.1002/mrd.23711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 09/06/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023]
Abstract
Although rat preimplantation embryos are necessary for producing genetically modified rats, their in vitro culture remains a challenge. Rat zygotes can develop from the one-cell stage to the blastocyst stage in vitro; however, long-term culture reduces their developmental competence via an unknown mechanism. In this study, we examined how in vitro conditions affect rat preimplantation embryos, which may explain this reduced competence. Comprehensive gene expression analysis showed that genes related to apoptosis and energy metabolism were differentially expressed in rat embryos cultured long-term in vitro compared with those developed in vivo. Furthermore, we found that the expression of Bak1 and Bax, which are responsible for mitochondrial outer membrane permeabilization, were more upregulated in embryos cultured in vitro than those developed in vivo. Similarly, apoptosis-dependent DNA fragmentation was also exacerbated in in vitro culture conditions. Finally, gene disruption using CRISPR/Cas9 showed that Bax, but not Bak1, was responsible for these effects. These findings suggest that long-term in vitro culture induces Bax-dependent apoptosis through the mitochondrial pathway and may provide clues to improve the long-term culture of rat preimplantation embryos for genetic engineering research.
Collapse
Affiliation(s)
- Kazuomi Nakamura
- Advanced Medicine, Innovation and Clinical Research Center, Tottori University Hospital, Yonago, Tottori, Japan
| | - Misako Seno
- Advanced Medicine & Translational Research Center, Organization for Research Initiative and Promotion, Tottori University, Yonago, Tottori, Japan
| | - Yuki Yoshimura
- Division of Integrative Physiology, Faculty of Medicine, Tottori University, Yonago, Tottori, Japan
| | - Osamu Suzuki
- Laboratory of Animal Models for Human Diseases, National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, Japan
| |
Collapse
|
7
|
Seki S, Kawabe T, Yamazaki W, Matsumura K, Oikawa T, Obata T, Higashiya M, Yano M, Eto T. Cryopreservation of rat embryos at all developmental stages by small-volume vitrification procedure and rapid warming in cryotubes. Sci Rep 2023; 13:20903. [PMID: 38017006 PMCID: PMC10684866 DOI: 10.1038/s41598-023-47394-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/13/2023] [Indexed: 11/30/2023] Open
Abstract
Intracellular ice formation during cryopreservation is lethal to the cell, including during warming. Here, we examined the effect of sample volume and warming rate on the cryopreservation success of 1-cell rat embryos based on successful development into blastocysts in vitro and to term in vivo following embryo transfer. Embryos were equilibrated in 5% propylene glycol solution for 10 min, held for 40 s at 0 °C in cryopreservation solution (5%PG + PEPeS), and cooled by immersion in liquid nitrogen. When 1-cell embryos were cryopreserved in a volume of 30-100 μL at a cooling rate of 5830-7160 °C/min and warmed at 35,480-49,400 °C/min by adding 1 mL of 0.3 M sucrose solution at 50 °C, 17.3-28.8% developed into blastocysts, compared with 57.0% of untreated embryos. However, when 1-cell embryos were cryopreserved in a smaller volume of 15 μl at 7950 °C/min and warmed at 68,850 °C/min, 58.8 ± 10.6% developed into blastocysts and 50.0 ± 7.4% developed to term, comparable to that of non-treated embryos (57.0 ± 5.4% and 51.4 ± 3.1%, respectively). Cryopreserved embryos at other developmental stages also showed high in vitro culture potential similar to that of the control. Using a conventional cryotube and a small-volume vitrification procedure with rapid warming, we achieved high levels of subsequent rat embryonic development at all developmental stages.
Collapse
Affiliation(s)
- Shinsuke Seki
- Experimental Animal Division, Bioscience Education and Research Support Center, Akita University, 1-1-1 Hondo, Akita, Akita, 010-8543, Japan.
| | - Toshiaki Kawabe
- ARK Resource Co., Ltd., 456 Osozu, Misato-machi, Shimomashiki-gun, Kumamoto, 861-4401, Japan
| | - Wataru Yamazaki
- Experimental Animal Division, Bioscience Education and Research Support Center, Akita University, 1-1-1 Hondo, Akita, Akita, 010-8543, Japan
| | - Kazuaki Matsumura
- School of Materials Science, Japan Advanced Institute of Science and Technology, 1-1 Asahi-dai, Nomi, Ishikawa, 923-1292, Japan
| | - Takanori Oikawa
- Experimental Animal Division, Bioscience Education and Research Support Center, Akita University, 1-1-1 Hondo, Akita, Akita, 010-8543, Japan
| | - Takahiro Obata
- Experimental Animal Division, Bioscience Education and Research Support Center, Akita University, 1-1-1 Hondo, Akita, Akita, 010-8543, Japan
| | - Misako Higashiya
- Experimental Animal Division, Bioscience Education and Research Support Center, Akita University, 1-1-1 Hondo, Akita, Akita, 010-8543, Japan
| | - Megumi Yano
- Experimental Animal Division, Bioscience Education and Research Support Center, Akita University, 1-1-1 Hondo, Akita, Akita, 010-8543, Japan
| | - Tomoo Eto
- Central Institute for Experimental Animals, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan.
| |
Collapse
|
8
|
Miyazawa Y, Fujinoki M. Enhancement of rat spermatozoal hyperactivation by progesterone. J Reprod Dev 2023; 69:279-290. [PMID: 37690839 PMCID: PMC10602764 DOI: 10.1262/jrd.2023-040] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 08/19/2023] [Indexed: 09/12/2023] Open
Abstract
Progesterone (P) is a well-known enhancer of hyperactivation which is associated with the success of in vitro fertilization (IVF). In this study, we examined whether P-enhanced hyperactivation affected IVF success in rats. When rat spermatozoa were exposed to 10, 20, and 40 ng/ml P, 20 ng/ml P enhanced hyperactivation via the membrane progesterone receptor. In addition, the enhancement of hyperactivation by 20 ng/ml P was regulated by phospholipase C, transmembrane adenylate cyclase, and protein kinase A. However, 20 ng/ml P did not affect IVF success. These results suggest that 20 ng/ml P enhances rat spermatozoal hyperactivation through non-genomic pathways. Because the concentration of P changes during the estrous cycle, it seems that rat spermatozoa are hyperactivated in response to the oviductal environment. However, the effect of 20 ng/ml P does not seem to fully capacitate spermatozoa.
Collapse
Affiliation(s)
- Yuki Miyazawa
- Research Laboratory of Laboratory Animals, Research Center for Laboratory Animals, Comprehensive Research Facilities for Advanced Medical Science, School of Medicine, Dokkyo Medical University, Tochigi 321-0293, Japan
| | - Masakatsu Fujinoki
- Research Laboratory of Laboratory Animals, Research Center for Laboratory Animals, Comprehensive Research Facilities for Advanced Medical Science, School of Medicine, Dokkyo Medical University, Tochigi 321-0293, Japan
| |
Collapse
|
9
|
Kobayashi K, Iwasa K, Azuma-Suzuki R, Kawauchi T, Nabeshima YI. Feto-maternal cholesterol transport regulated by β-Klotho-FGF15 axis is essential for fetal growth. Life Sci Alliance 2023; 6:e202301916. [PMID: 37541847 PMCID: PMC10403640 DOI: 10.26508/lsa.202301916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/06/2023] Open
Abstract
β-Klotho (β-KL) is indispensable to regulate lipid, glucose, and energy metabolism in adult animals. β-KL is highly expressed in the yolk sac, but its role in the developmental stages has not been established. We hypothesized that β-KL is required for metabolic regulation in the embryo and aimed to clarify the role of β-KL during development. Here, we show that β-KL regulates feto-maternal cholesterol transport through the yolk sac by mediating FGF 15 signaling, and also that impairment of the β-KL-FGF15 axis causes fetal growth restriction (FGR). Embryos of β- kl knockout (β-kl-/-) mice were morphologically normal but exhibited FGR before placental maturation. The body weight of β-kl-/- mice remained lower after birth. β-KL deletion reduced cholesterol supply from the maternal blood and led to lipid shortage in the embryos. These phenotypes were similar to those of embryos lacking FGF15, indicating that β-KL-FGF15 axis is essential for growth and lipid regulation in the embryonic stages. Our findings suggest that lipid abnormalities in early gestation provoke FGR, leading to reduced body size in later life.
Collapse
Affiliation(s)
- Kanako Kobayashi
- Department of Aging Science and Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Kazuko Iwasa
- Department of Aging Science and Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Rika Azuma-Suzuki
- Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| | - Takeshi Kawauchi
- Department of Aging Science and Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
- Department of Adaptive and Maladaptive Responses in Health and Disease, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Yo-Ichi Nabeshima
- Department of Aging Science and Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| |
Collapse
|
10
|
Kageyama A, Tsuchiya M, Terakawa J, Ito J, Kashiwazaki N. A combined treatment with progesterone, anti-inhibin serum, and equine chorionic gonadotropin improves number of ovulated oocytes in young C57BL/6J mice. J Reprod Dev 2023; 69:223-226. [PMID: 37331813 PMCID: PMC10435527 DOI: 10.1262/jrd.2023-036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/24/2023] [Indexed: 06/20/2023] Open
Abstract
Superovulation procedures are routinely and widely used in mouse reproductive technology. Previous studies have shown that a large number of oocytes can be obtained from adult mice (> 10 weeks old) using a combined treatment with progesterone (P4) and anti-inhibin serum (AIS). However, these effects have not been fully investigated in young (4 weeks) C57BL/6J mice. Here, we found that a modified superovulation protocol (combined treatment with P4, AIS, eCG (equine chorionic gonadotropin), and hCG (human chorionic gonadotropin); P4D2-Ae-h) improved the number of oocytes compared to the control (eCG and hCG) (39.7 vs. 21.3 oocytes/mouse). After in vitro fertilization, pronuclear formation rates were 69.3% (P4D2-Ae-h group) and 66.2% (control group). After embryo transfer, 46.4% (116/250) of the embryos in the P4D2-Ae-h group successfully developed to term, which was comparable to the control group (42.9%; 123/287 embryos). In conclusion, our protocol (P4D2-Ae-h) was effective for superovulation in young C57BL/6J mice.
Collapse
Affiliation(s)
- Atsuko Kageyama
- Laboratory of Animal Reproduction, Graduate School of Veterinary Sciences, Azabu University, Sagamihara 252-5201, Japan
| | - Mizuho Tsuchiya
- School of Veterinary Medicine, Azabu University, Sagamihara 252-5201, Japan
| | - Jumpei Terakawa
- Laboratory of Toxicology, School of Veterinary Medicine, Azabu University, Sagamihara 252-5201, Japan
| | - Junya Ito
- Laboratory of Animal Reproduction, Graduate School of Veterinary Sciences, Azabu University, Sagamihara 252-5201, Japan
- School of Veterinary Medicine, Azabu University, Sagamihara 252-5201, Japan
- Center for Human and Animal Symbiosis Science, Azabu University, Sagamihara 252-5201, Japan
| | - Naomi Kashiwazaki
- Laboratory of Animal Reproduction, Graduate School of Veterinary Sciences, Azabu University, Sagamihara 252-5201, Japan
- School of Veterinary Medicine, Azabu University, Sagamihara 252-5201, Japan
| |
Collapse
|
11
|
Kano M, Mizuno N, Sato H, Kimura T, Hirochika R, Iwasaki Y, Inoshita N, Nagano H, Kasai M, Yamamoto H, Yamaguchi T, Suga H, Masaki H, Mizutani E, Nakauchi H. Functional calcium-responsive parathyroid glands generated using single-step blastocyst complementation. Proc Natl Acad Sci U S A 2023; 120:e2216564120. [PMID: 37379351 PMCID: PMC10334775 DOI: 10.1073/pnas.2216564120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 04/24/2023] [Indexed: 06/30/2023] Open
Abstract
Patients with permanent hypoparathyroidism require lifelong replacement therapy to avoid life-threatening complications, The benefits of conventional treatment are limited, however. Transplanting a functional parathyroid gland (PTG) would yield better results. Parathyroid gland cells generated from pluripotent stem cells in vitro to date cannot mimic the physiological responses to extracellular calcium that are essential for calcium homeostasis. We thus hypothesized that blastocyst complementation (BC) could be a better strategy for generating functional PTG cells and compensating loss of parathyroid function. We here describe generation of fully functional PTGs from mouse embryonic stem cells (mESCs) with single-step BC. Using CRISPR-Cas9 knockout of Glial cells missing2 (Gcm2), we efficiently produced aparathyroid embryos for BC. In these embryos, mESCs differentiated into endocrinologically mature PTGs that rescued Gcm2-/- mice from neonatal death. The mESC-derived PTGs responded to extracellular calcium, restoring calcium homeostasis on transplantation into mice surgically rendered hypoparathyroid. We also successfully generated functional interspecies PTGs in Gcm2-/- rat neonates, an accomplishment with potential for future human PTG therapy using xenogeneic animal BC. Our results demonstrate that BC can produce functional endocrine organs and constitute a concept in treatment of hypoparathyroidism.
Collapse
Affiliation(s)
- Mayuko Kano
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo113-8510, Japan
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo108-8639, Japan
- Metabolism and Endocrinology, Department of Medicine, St. Marianna University School of Medicine, Miyamae-ku, Kawasaki, Kanagawa216-8511, Japan
| | - Naoaki Mizuno
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo113-8510, Japan
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo108-8639, Japan
| | - Hideyuki Sato
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo113-8510, Japan
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo108-8639, Japan
| | - Takaharu Kimura
- Laboratory of Stem Cell Therapy, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki305-8577, Japan
| | - Rei Hirochika
- Laboratory of Stem Cell Therapy, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki305-8577, Japan
| | - Yasumasa Iwasaki
- Department of Clinical Nutrition, Faculty of Health Science, Suzuka University of Medical Science, Suzuka, Mie510-0293, Japan
- Department of Endocrinology, Metabolism, and Nephrology, Kochi Medical School, Kochi University, Oko-cho, Nankoku, Kochi783-8505, Japan
| | - Naoko Inoshita
- Department of Pathology, Moriyama Memorial Hospital, Edogawa-ku, Tokyo134-0081, Japan
| | - Hisato Nagano
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo113-8510, Japan
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo108-8639, Japan
- Department of Plastic and Reconstructive Surgery, National Defense Medical College, Tokorozawa, Saitama359-8513, Japan
| | - Mariko Kasai
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo113-8510, Japan
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo108-8639, Japan
| | - Hiromi Yamamoto
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo113-8510, Japan
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo108-8639, Japan
| | - Tomoyuki Yamaguchi
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo108-8639, Japan
- Laboratory of Regenerative Medicine, Tokyo University of Pharmacy and Life Science, Hachioji, Tokyo192-0392, Japan
| | - Hidetaka Suga
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya466-8550, Japan
| | - Hideki Masaki
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo113-8510, Japan
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo108-8639, Japan
| | - Eiji Mizutani
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo113-8510, Japan
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo108-8639, Japan
- Laboratory of Stem Cell Therapy, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki305-8577, Japan
| | - Hiromitsu Nakauchi
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University (TMDU), Bunkyo-ku, Tokyo113-8510, Japan
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo108-8639, Japan
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA94305
| |
Collapse
|
12
|
Morita K, Honda A, Asano M. A Simple and Efficient Method for Generating KO Rats Using In Vitro Fertilized Oocytes. Methods Mol Biol 2023; 2637:233-246. [PMID: 36773151 DOI: 10.1007/978-1-0716-3016-7_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
The development of ZFN, TALEN, and CRISPR/Cas9 systems has simplified the process of generating knockout (KO) and knock-in (KI) rats in addition to mice. However, in rats, an efficient genome editing technique that uses in vitro fertilized oocytes has not been established. Recently, we reported the stable generation of offspring from five standard strains of rats by superovulation and in vitro fertilization (IVF). Furthermore, genome-edited rats can be easily generated by electroporation. First, juvenile female rats are administered LHRH (luteinizing hormone-releasing hormone) to synchronize the estrous cycle and then AIS (Automatic Identification System) with PMSG (pregnant mare serum gonadotropin) before hCG (human chorionic gonadotropin) for superovulation. Sperm collected from a sexually mature male rat the following morning is then pre-cultured. Cumulus cell-oocyte complexes (COCs) are collected from female rats under anesthesia, and COCs are induced into a medium containing concentration-adjusted sperm. Thereafter, oocytes with two pronucleus are selected as fertilized oocytes. Next, fertilized oocytes are transferred into a glass chamber containing CRISPR ribonucleoprotein (RNP) complexes formed from gRNA and Cas9 protein. After electroporation, fertilized oocytes are then immediately transferred to culture medium. The next day, embryos are transferred into the oviduct of pseudopregnant female rats. Using the above method, offspring can be obtained 22 days after the day of embryo transfer. In this paper, we outline a method allowing simple and efficient generation of genetically modified rats without the need for technically difficult micromanipulation techniques.
Collapse
Affiliation(s)
- Kohtaro Morita
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Arata Honda
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Jichi Medical University, School of Medicine, Tochigi, Japan
- RIKEN BioResource Research Center, Tsukuba, Ibaraki, Japan
| | - Masahide Asano
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
13
|
A novel technique for large-fragment knock-in animal production without ex vivo handling of zygotes. Sci Rep 2023; 13:2245. [PMID: 36755180 PMCID: PMC9908863 DOI: 10.1038/s41598-023-29468-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 02/06/2023] [Indexed: 02/10/2023] Open
Abstract
CRISPR/Cas-based genome editing has dramatically improved genetic modification technology. In situ electroporation called genome editing via oviductal nucleic acid delivery (GONAD), which eliminates the need for ex vivo embryo handling, is technically the simplest method for gene transfer and can be performed in laboratories without developmental engineering expertise including micromanipulation techniques. However, the use of this method remains challenging in the case of large-fragment knock-in, such as gene expression cassettes. Adeno-associated viruses (AAV) act as donor DNA for homologous recombination in infected cells, including rodent embryos. In this study, we demonstrated simultaneous electroporation of AAV donors and CRISPR/Cas9 components into embryos to create knock-in animals, and successfully generated knock-in rats carrying a gene cassette with a length of 3.0 kb using a small number of animals and in situ electroporation. These findings indicate that this technique is an efficient high-throughput strategy for producing genetically modified rodents and may be applicable to other animal species.
Collapse
|
14
|
TORIKAI K, SHIMIZU K, NAGATOMO H, KASAI M, KATO-ITOH M, KAMADA Y, SHIBASAKI I, JEON H, KIKUCHI R, WAKAYAMA S, SUCHY F, NAKAUCHI H, WAKAYAMA T, MIZUTANI E. Removal of sperm tail using trypsin and pre-activation of oocyte facilitates intracytoplasmic sperm injection in mice and rats. J Reprod Dev 2023; 69:48-52. [PMID: 36529517 PMCID: PMC9939287 DOI: 10.1262/jrd.2022-065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
We examined various methods to enhance the accessibility of intracytoplasmic sperm injection (ICSI) technology to more users by making the technique easier, more efficient, and practical. First, the methods for artificially removing the mouse sperm tail were evaluated. Trypsin treatment was found to efficiently remove the sperm tails. The resultant sperm cells had a lower oocyte activation capacity; however, the use of activated oocytes resulted in the same fecundity as that of fresh, untreated sperm. Pre-activated oocytes were more resistant to physical damage, showed higher survival rates, and required less time per injection. Testing this method in rats yielded similar results, although the oocyte activation method was different. Remarkably, this method resulted in higher birth rates of rat progeny than with conventional methods of rat ICSI. Our method thereby streamlines mouse and rat ICSI, making it more accessible to laboratories across many disciplines.
Collapse
Affiliation(s)
- Kohei TORIKAI
- Faculty of Life and Environmental Sciences, University of Yamanashi, Yamanashi 400-8510, Japan
| | - Kazuma SHIMIZU
- Faculty of Life and Environmental Sciences, University of Yamanashi, Yamanashi 400-8510, Japan
| | - Hiroaki NAGATOMO
- Center for Life Science Research, University of Yamanashi, Yamanashi, 409-3898, Japan
| | - Mariko KASAI
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Megumi KATO-ITOH
- Division of Stem Cell Therapy, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Yuko KAMADA
- Faculty of Life and Environmental Sciences, University of Yamanashi, Yamanashi 400-8510, Japan
| | - Ikue SHIBASAKI
- Faculty of Life and Environmental Sciences, University of Yamanashi, Yamanashi 400-8510, Japan
| | - Hyojung JEON
- Laboratory of Stem Cell Therapy, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Riko KIKUCHI
- Laboratory of Stem Cell Therapy, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Sayaka WAKAYAMA
- Advanced Biotechnology Center, University of Yamanashi, Yamanashi 400-8510, Japan
| | - Fabian SUCHY
- Institute for Stem Cell Biology and Regenerative Medicine, Department of Genetics, Stanford University School of Medicine, California 94305, USA
| | - Hiromitsu NAKAUCHI
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University, Tokyo 113-8510, Japan,Institute for Stem Cell Biology and Regenerative Medicine, Department of Genetics, Stanford University School of Medicine, California 94305, USA
| | - Teruhiko WAKAYAMA
- Faculty of Life and Environmental Sciences, University of Yamanashi, Yamanashi 400-8510, Japan,Advanced Biotechnology Center, University of Yamanashi, Yamanashi 400-8510, Japan
| | - Eiji MIZUTANI
- Stem Cell Therapy Laboratory, Advanced Research Institute, Tokyo Medical and Dental University, Tokyo 113-8510, Japan,Laboratory of Stem Cell Therapy, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
15
|
Sari GP, Hilario PLL, Yuri S, Honda A, Isotani A. Scheduled simple production method of pseudopregnant female mice for embryo transfer using the luteinizing hormone-releasing hormone agonist. Sci Rep 2022; 12:21985. [PMID: 36539541 PMCID: PMC9767918 DOI: 10.1038/s41598-022-26425-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
The use of mice as experimental animal models has been a practice since the development of genetically engineered mouse models (GEMMs) in the early 1980s. New technologies, including genome editing, have helped in the time- and cost-efficient generation of GEMMs. However, methods for preparing pseudopregnant females, essential for the generation of GEMMs, remain less advanced. This study proposes a new method to achieve simple production of pseudopregnant female mice using a luteinizing hormone-releasing hormone agonist (LHRHa). A 20 µg LHRHa/mouse was identified as the best dose for inducing estrus synchronization. However, the frequency of copulation was 40% on a single injection. With sequential injections of 20 µg LHRHa/mouse on Days-1 and -2, followed by pairing on Day-5, 74% of LHRHa-treated females copulated with male mice. Moreover, LHRHa treatment did not affect fetal and postnatal development. Eventually, successful generation of offspring via embryo transfer was attained using LHRHa-treated pseudopregnant females. LHRHa administration method is efficient in producing pseudopregnant female mice for the generation of GEMMs, and we expect that it will contribute towards advancing the clinical research.
Collapse
Affiliation(s)
- Gema Puspa Sari
- grid.260493.a0000 0000 9227 2257Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama-Cho, Ikoma, Nara 630-0192 Japan
| | - Patrick Louis Lagman Hilario
- grid.260493.a0000 0000 9227 2257Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama-Cho, Ikoma, Nara 630-0192 Japan
| | - Shunsuke Yuri
- grid.260493.a0000 0000 9227 2257Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama-Cho, Ikoma, Nara 630-0192 Japan
| | - Arata Honda
- grid.410804.90000000123090000Center for Development of Advanced Medical Technology, School of Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-Shi, Tochigi-Ken 329-0498 Japan
| | - Ayako Isotani
- grid.260493.a0000 0000 9227 2257Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama-Cho, Ikoma, Nara 630-0192 Japan
| |
Collapse
|
16
|
Takeo T, Nakao S, Mikoda N, Yamaga K, Maeda R, Tsuchiyama S, Nakatsukasa E, Nakagata N. Optimized protocols for sperm cryopreservation and in vitro fertilization in the rat. Lab Anim (NY) 2022; 51:256-274. [DOI: 10.1038/s41684-022-01053-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 08/15/2022] [Indexed: 11/05/2022]
|
17
|
Sato M, Nakamura S, Inada E, Takabayashi S. Recent Advances in the Production of Genome-Edited Rats. Int J Mol Sci 2022; 23:ijms23052548. [PMID: 35269691 PMCID: PMC8910656 DOI: 10.3390/ijms23052548] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/17/2022] [Accepted: 02/21/2022] [Indexed: 12/14/2022] Open
Abstract
The rat is an important animal model for understanding gene function and developing human disease models. Knocking out a gene function in rats was difficult until recently, when a series of genome editing (GE) technologies, including zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and the type II bacterial clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated Cas9 (CRISPR/Cas9) systems were successfully applied for gene modification (as exemplified by gene-specific knockout and knock-in) in the endogenous target genes of various organisms including rats. Owing to its simple application for gene modification and its ease of use, the CRISPR/Cas9 system is now commonly used worldwide. The most important aspect of this process is the selection of the method used to deliver GE components to rat embryos. In earlier stages, the microinjection (MI) of GE components into the cytoplasm and/or nuclei of a zygote was frequently employed. However, this method is associated with the use of an expensive manipulator system, the skills required to operate it, and the egg transfer (ET) of MI-treated embryos to recipient females for further development. In vitro electroporation (EP) of zygotes is next recognized as a simple and rapid method to introduce GE components to produce GE animals. Furthermore, in vitro transduction of rat embryos with adeno-associated viruses is potentially effective for obtaining GE rats. However, these two approaches also require ET. The use of gene-engineered embryonic stem cells or spermatogonial stem cells appears to be of interest to obtain GE rats; however, the procedure itself is difficult and laborious. Genome-editing via oviductal nucleic acids delivery (GONAD) (or improved GONAD (i-GONAD)) is a novel method allowing for the in situ production of GE zygotes existing within the oviductal lumen. This can be performed by the simple intraoviductal injection of GE components and subsequent in vivo EP toward the injected oviducts and does not require ET. In this review, we describe the development of various approaches for producing GE rats together with an assessment of their technical advantages and limitations, and present new GE-related technologies and current achievements using those rats in relation to human diseases.
Collapse
Affiliation(s)
- Masahiro Sato
- Department of Genome Medicine, National Center for Child Health and Development, Tokyo 157-8535, Japan
- Correspondence: (M.S.); (S.T.); Tel.: +81-3-3416-0181 (M.S.); +81-53-435-2001 (S.T.)
| | - Shingo Nakamura
- Division of Biomedical Engineering, National Defense Medical College Research Institute, Saitama 359-8513, Japan;
| | - Emi Inada
- Department of Pediatric Dentistry, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan;
| | - Shuji Takabayashi
- Laboratory Animal Facilities & Services, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
- Correspondence: (M.S.); (S.T.); Tel.: +81-3-3416-0181 (M.S.); +81-53-435-2001 (S.T.)
| |
Collapse
|
18
|
Matsumura T, Noda T, Satouh Y, Morohoshi A, Yuri S, Ogawa M, Lu Y, Isotani A, Ikawa M. Sperm IZUMO1 Is Required for Binding Preceding Fusion With Oolemma in Mice and Rats. Front Cell Dev Biol 2022; 9:810118. [PMID: 35096839 PMCID: PMC8790511 DOI: 10.3389/fcell.2021.810118] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 12/21/2021] [Indexed: 01/28/2023] Open
Abstract
Fertilization occurs as the culmination of multi-step complex processes. First, mammalian spermatozoa undergo the acrosome reaction to become fusion-competent. Then, the acrosome-reacted spermatozoa penetrate the zona pellucida and adhere to and finally fuse with the egg plasma membrane. IZUMO1 is the first sperm protein proven to be essential for sperm-egg fusion in mammals, as Izumo1 knockout mouse spermatozoa adhere to but fail to fuse with the oolemma. However, the IZUMO1 function in other species remains largely unknown. Here, we generated Izumo1 knockout rats by CRISPR/Cas9 and found the male rats were infertile. Unlike in mice, Izumo1 knockout rat spermatozoa failed to bind to the oolemma. Further investigation revealed that the acrosome-intact sperm binding conceals a decreased number of the acrosome-reacted sperm bound to the oolemma in Izumo1 knockout mice. Of note, we could not see any apparent defects in the binding of the acrosome-reacted sperm to the oolemma in the mice lacking recently found fusion-indispensable genes, Fimp, Sof1, Spaca6, or Tmem95. Collectively, our data suggest that IZUMO1 is required for the sperm-oolemma binding prior to fusion at least in rat.
Collapse
Affiliation(s)
- Takafumi Matsumura
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Taichi Noda
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Division of Reproductive Biology, Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
- Priority Organization for Innovation and Excellence, Kumamoto University, Kumamoto, Japan
| | - Yuhkoh Satouh
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Akane Morohoshi
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Graduate School of Medicine, Osaka University, Suita, Japan
| | - Shunsuke Yuri
- Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Japan
| | - Masaki Ogawa
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Yonggang Lu
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Ayako Isotani
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Japan
- Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Masahito Ikawa
- Department of Experimental Genome Research, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
- Graduate School of Medicine, Osaka University, Suita, Japan
- Immunology Frontier Research Center, Osaka University, Suita, Japan
- Laboratory of Reproductive Systems Biology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
19
|
Miura K, Ogura A, Kobatake K, Honda H, Kaminuma O. Progress of genome editing technology and developmental biology useful for radiation research. JOURNAL OF RADIATION RESEARCH 2021; 62:i53-i63. [PMID: 33978171 PMCID: PMC8114227 DOI: 10.1093/jrr/rraa127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 11/26/2020] [Accepted: 11/23/2020] [Indexed: 06/12/2023]
Abstract
Following the development of genome editing technology, it has become more feasible to create genetically modified animals such as knockout (KO), knock-in, and point-mutated animals. The genome-edited animals are useful to investigate the roles of various functional genes in many fields of biological science including radiation research. Nevertheless, some researchers may experience difficulty in generating genome-edited animals, probably due to the requirement for equipment and techniques for embryo manipulation and handling. Furthermore, after obtaining F0 generation, genome-edited animals generally need to be expanded and maintained for analyzing the target gene function. To investigate genes essential for normal birth and growth, the generation of conditional KO (cKO) animals in which a tissue- or stage-specific gene mutation can be introduced is often required. Here, we describe the basic principle and application of genome editing technology including zinc-finger nuclease, transcription-activator-like effector nuclease, and clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR associated protein (Cas) systems. Recently advanced developmental biology methods have enabled application of the technology, especially CRISPR/Cas, to zygotes, leading to the prompt production of genome-edited animals. For pre-implantation embryos, genome editing via oviductal nucleic acid delivery has been developed as an embryo manipulation- or handling-free method. Examining the gene function at F0 generation is becoming possible by employing triple-target CRISPR technology. This technology, in combination with a blastocyst complementation method enables investigation of even birth- and growth-responsible genes without establishing cKO strains. We hope that this review is helpful for understanding and expanding genome editing-related technology and for progressing radiation research.
Collapse
Affiliation(s)
- Kento Miura
- Department of Disease Model, Research Institute of Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8553, Japan
- RIKEN BioResource Research Center, Tsukuba, Ibaraki 305-0074, Japan
| | - Atsuo Ogura
- RIKEN BioResource Research Center, Tsukuba, Ibaraki 305-0074, Japan
- RIKEN Cluster for Pioneering Research, Wako, Saitama 351-0198, Japan
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8572, Japan
| | - Kohei Kobatake
- Department of Disease Model, Research Institute of Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8553, Japan
- Department of Urology, Hiroshima University, Hiroshima 734-8553, Japan
| | - Hiroaki Honda
- Field of Human Disease Models, Major in Advanced Life Sciences and Medicine, Institute of Laboratory Animals, Tokyo Women's Medical University, Tokyo 162-8666, Japan
| | - Osamu Kaminuma
- Department of Disease Model, Research Institute of Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8553, Japan
- RIKEN BioResource Research Center, Tsukuba, Ibaraki 305-0074, Japan
| |
Collapse
|
20
|
Hart-Johnson S, Mankelow K. Archiving genetically altered animals: a review of cryopreservation and recovery methods for genome edited animals. Lab Anim 2021; 56:26-34. [PMID: 33847177 DOI: 10.1177/00236772211007306] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
With the ever-expanding numbers of genetically altered (GA) animals created in this new age of CRISPR/Cas, tools for helping the management of this vast and valuable resource are essential. Cryopreservation of embryos and germplasm of GA animals has been a widely used tool for many years now, allowing for the archiving, distribution and colony management of stock. However, each year brings an array of advances, improving survival rates of embryos, success rates of in-vitro fertilisation and the ability to better share lines and refine the methods to preserve them. This article will focus on the mouse field, referencing the latest developments and assessing their efficacy and ease of implementation, with a brief note on other common genetically altered species (rat, zebrafish, Xenopus, avian species and non-human Primates).
Collapse
|
21
|
Abstract
Genetically modified animals, especially rodents, are widely used in biomedical research. However, non-rodent models are required for efficient translational medicine and preclinical studies. Owing to the similarity in the physiological traits of pigs and humans, genetically modified pigs may be a valuable resource for biomedical research. Somatic cell nuclear transfer (SCNT) using genetically modified somatic cells has been the primary method for the generation of genetically modified pigs. However, site-specific gene modification in porcine cells is inefficient and requires laborious and time-consuming processes. Recent improvements in gene-editing systems, such as zinc finger nucleases, transcription activator-like effector nucleases, and the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein (CRISPR/Cas) system, represent major advances. The efficient introduction of site-specific modifications into cells via gene editors dramatically reduces the effort and time required to generate genetically modified pigs. Furthermore, gene editors enable direct gene modification during embryogenesis, bypassing the SCNT procedure. The application of gene editors has progressively expanded, and a range of strategies is now available for porcine gene engineering. This review provides an overview of approaches for the generation of genetically modified pigs using gene editors, and highlights the current trends, as well as the limitations, of gene editing in pigs.
Collapse
Affiliation(s)
- Fuminori Tanihara
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima 770-8513, Japan.,Center for Development of Advanced Medical Technology, Jichi Medical University, Tochigi 329-0498, Japan
| | - Maki Hirata
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima 770-8513, Japan
| | - Takeshige Otoi
- Faculty of Bioscience and Bioindustry, Tokushima University, Tokushima 770-8513, Japan
| |
Collapse
|
22
|
One-Step Generation of Multiple Gene-Edited Pigs by Electroporation of the CRISPR/Cas9 System into Zygotes to Reduce Xenoantigen Biosynthesis. Int J Mol Sci 2021; 22:ijms22052249. [PMID: 33668187 PMCID: PMC7956194 DOI: 10.3390/ijms22052249] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/18/2021] [Accepted: 02/20/2021] [Indexed: 02/08/2023] Open
Abstract
Xenoantigens cause hyperacute rejection and limit the success of interspecific xenografts. Therefore, genes involved in xenoantigen biosynthesis, such as GGTA1, CMAH, and B4GALNT2, are key targets to improve the outcomes of xenotransplantation. In this study, we introduced a CRISPR/Cas9 system simultaneously targeting GGTA1, CMAH, and B4GALNT2 into in vitro-fertilized zygotes using electroporation for the one-step generation of multiple gene-edited pigs without xenoantigens. First, we optimized the combination of guide RNAs (gRNAs) targeting GGTA1 and CMAH with respect to gene editing efficiency in zygotes, and transferred electroporated embryos with the optimized gRNAs and Cas9 into recipient gilts. Next, we optimized the Cas9 protein concentration with respect to the gene editing efficiency when GGTA1, CMAH, and B4GALNT2 were targeted simultaneously, and generated gene-edited pigs using the optimized conditions. We achieved the one-step generation of GGTA1/CMAH double-edited pigs and GGTA1/CMAH/B4GALNT2 triple-edited pigs. Immunohistological analyses demonstrated the downregulation of xenoantigens; however, these multiple gene-edited pigs were genetic mosaics that failed to knock out some xenoantigens. Although mosaicism should be resolved, the electroporation technique could become a primary method for the one-step generation of multiple gene modifications in pigs aimed at improving pig-to-human xenotransplantation.
Collapse
|
23
|
Zhang C, Ren Z, Gong Z. Transgenic Expression and Genome Editing by Electroporation of Zebrafish Embryos. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2020; 22:644-650. [PMID: 32748174 DOI: 10.1007/s10126-020-09985-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/21/2020] [Indexed: 05/22/2023]
Abstract
Microinjection is predominantly used to produce genetically modified fish. However, there are certain difficulties involved in some fish species. In this study, we tested an alternative method to produce genetically modified zebrafish by delivering DNA and other materials into embryos by electroporation. We optimized the electroporation conditions of a square wave electroporation system that work efficiently for the introduction of plasmid DNA, recombinant Cas9 nuclease and synthetic dual guide RNAs. Transgenic expression was observed in a wide range of tissues, which is comparable with those obtained by microinjection. We further determined that efficient gene delivery can be achieved during the cleavage stage. This study describes detailed electroporation parameters for gene delivery with high efficiency and low toxicity, providing a novel method to generate transgenic lines and genome editing.
Collapse
Affiliation(s)
- Changqing Zhang
- Department of Biological Sciences, National University of Singapore, 14 Sciences Drive 4, Singapore, 117558, Singapore
| | - Ziheng Ren
- Department of Biological Sciences, National University of Singapore, 14 Sciences Drive 4, Singapore, 117558, Singapore
| | - Zhiyuan Gong
- Department of Biological Sciences, National University of Singapore, 14 Sciences Drive 4, Singapore, 117558, Singapore.
| |
Collapse
|
24
|
Hino C, Ueda J, Funakoshi H, Matsumoto S. Defined oocyte collection time is critical for reproducible in vitro fertilization in rats of different strains. Theriogenology 2020; 144:146-151. [PMID: 31940506 DOI: 10.1016/j.theriogenology.2020.01.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 12/01/2019] [Accepted: 01/05/2020] [Indexed: 11/24/2022]
Abstract
In vitro fertilization (IVF) is an established technology that is widely used in reproductive engineering. However, in rats, successful application of IVF is difficult to achieve, and it has had poor reproducibility. In a previous study on the critical issues associated with successful IVF in Wistar rats, we investigated the influence of oocyte collection duration on fertilization rates by dividing the procedure into three steps (oviduct extraction from euthanized animals, oocyte collection from the ampullae of oviducts, and oocyte preincubation until insemination), and identified the appropriate times for each. Here we show that use of the same defined duration for oviduct extraction from superovulated Wistar rats and for oocyte collection from the oviducts also produced highly reproducible fertilization rates of more than 90% in other rat strains. Furthermore, the versatility of these criteria was demonstrated using another IVF protocol. Thus, this simple procedure has enabled the standardization of IVF in rats and will enhance further experimental studies.
Collapse
Affiliation(s)
- Chihiro Hino
- Center for Advanced Research and Education, Asahikawa Medical University, Asahikawa, Japan.
| | - Jun Ueda
- Center for Advanced Research and Education, Asahikawa Medical University, Asahikawa, Japan.
| | - Hiroshi Funakoshi
- Center for Advanced Research and Education, Asahikawa Medical University, Asahikawa, Japan
| | - Seiji Matsumoto
- Center for Advanced Research and Education, Asahikawa Medical University, Asahikawa, Japan
| |
Collapse
|
25
|
Nakagata N, Mikoda N, Nakao S, Nakatsukasa E, Takeo T. Establishment of sperm cryopreservation and in vitro fertilisation protocols for rats. Sci Rep 2020; 10:93. [PMID: 31919412 PMCID: PMC6952447 DOI: 10.1038/s41598-019-57090-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 12/20/2019] [Indexed: 11/10/2022] Open
Abstract
Recently, genome-editing tools have come into common use in the field of rat research, and consequently, many genetically modified rat strains have been preserved and archived as frozen embryos. Although there have been many reports published on the topic of rat sperm cryopreservation, no report has yet provided satisfactory and acceptable protocols for the cryopreservation of rat sperm. In this study, we developed methods for both the cryopreservation of transgenic rat sperm and in vitro fertilisation using frozen sperm, which yielded high fertilisation rates.
Collapse
Affiliation(s)
- Naomi Nakagata
- Center for Animal Resources and Development, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan.
| | - Nobuyuki Mikoda
- Center for Animal Resources and Development, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Satohiro Nakao
- Center for Animal Resources and Development, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Ena Nakatsukasa
- Department of Animal Model Development, Brain Research Institute, Niigata University, 1-757 Asahimachidori, Chuo-ku, Niigata, 951-8585, Japan
| | - Toru Takeo
- Center for Animal Resources and Development, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| |
Collapse
|