Minireviews Open Access
Copyright ©The Author(s) 2015. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Stem Cells. Aug 26, 2015; 7(7): 1054-1063
Published online Aug 26, 2015. doi: 10.4252/wjsc.v7.i7.1054
Rat embryonic stem cells create new era in development of genetically manipulated rat models
Kazushi Kawaharada, Research and Development Division, DS Pharma Biomedical Co. Ltd., Osaka 564-0053, Japan
Masaki Kawamata, Takahiro Ochiya, Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo 104-0045, Japan
Author contributions: Kawaharada K, Kawamata M and Ochiya T contributed to this paper.
Conflict-of-interest statement: Kazushi Kawaharada is an employee of DS Pharma Biomedical Co., Ltd. The other authors declared no conflict of interest.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Dr. Takahiro Ochiya, Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan. tochiya@ncc.go.jp
Telephone: +81-3-35422511 Fax: +81-3-55650727
Received: August 22, 2014
Peer-review started: August 23, 2014
First decision: September 28, 2014
Revised: May 29, 2015
Accepted: July 16, 2015
Article in press: July 18, 2015
Published online: August 26, 2015

Abstract

Embryonic stem (ES) cells are isolated from the inner cell mass of a blastocyst, and are used for the generation of gene-modified animals. In mice, the transplantation of gene-modified ES cells into recipient blastocysts leads to the creation of gene-targeted mice such as knock-in and knock-out mice; these gene-targeted mice contribute greatly to scientific development. Although the rat is considered a useful laboratory animal alongside the mouse, fewer gene-modified rats have been produced due to the lack of robust establishment methods for rat ES cells. A new method for establishing rat ES cells using signaling inhibitors was reported in 2008. By considering the characteristics of rat ES cells, recent research has made progress in improving conditions for the stable culture of rat ES cells in order to generate gene-modified rats efficiently. In this review, we summarize several advanced methods to maintain rat ES cells and generate gene-targeted rats.

Key Words: Embryonic stem cells, Transgenic rat

Core tip: Rat embryonic stem (ES) cells are thought to be an essential tool for creating transgenic rats. Since the method for establishing rat ES cells using signaling inhibitors was reported, numerous approaches have been made to propagate rat ES cells efficiently. Additionally, recent investigations have demonstrated the usefulness of the signal inhibitors for microinjection. In this review, we summarize the several advanced methods to maintain rat ES cells and generate gene-targeted rats.



INTRODUCTION

Rattus norvegicus, is widely used as a laboratory animal in many kinds of scientific field, such as biochemical, biomedical, and pharmacological studies. In 1828, albino mutants were brought into laboratories for physiological studies such as fasting studies. Since the nineteenth century, over 700 rat strains including inbred and outbred strains have been developed, and have been used for a multitude of studies[1,2]. Rats offer some advantages over mice in some transplantation, behavior, and pharmacokinetic studies, because the larger size and greater intelligence of rats compared to mice enables ease of surgical operation, a large-volume of blood sampling, and assessment of high-level learning[3,4].

In addition, it has been suggested that rats are a useful model for physiological studies owing to the heart rate of rats being closer than that of mice to the human heart rate[2]. For these reasons, the publications involving rats outnumbered those involving mice for many years. However, far fewer scientific procedures have used transgenic rats than have used mice[5]. One of the reasons for this disparity has been a lack of reliable methods to establish rat embryonic stem (ES) cells. Genetically modified mice have been routinely created using mouse ES cells with gene-manipulation technologies. These genetically modified mice have contributed greatly to scientific development since the first establishment of mouse ES cell technology in 1981[6]. Meanwhile in rats, advancement of one of the gene-targeting technology using zinc-finger nucleases (ZFNs) allowed to generate the first knock-out rats in 2009[7,8]. Microinjection of ZFNs into the pronuclei of rat embryos leads to the creation of knock-out rats. ZFNs are engineered proteins with DNA-binding and nuclease activity, which facilitates the targeted editing of genomes by creating double-strand breaks in the DNA at specified locations and promoting non-homologous end-joining. Moreover, a robust method to establish rat ES cells was reported in 2008[9,10], and the generation of knock-out rats was achieved using rat ES cell-based technology in 2010[11].

ESTBLISHMENT, CHARACTERIZATION, AND MAINTENANCE OF RAT EMBRYONIC STEM CELLS
Attempts for establishment of rat embryonic stem cells

Rat ES cells are isolated from the inner cell mass (ICM) of a blastocyst. Figure 1 shows a procedure for the establishment of rat ES cells (our unpublished data). The defining properties of ES cells are following; they are derived from an ICM with pluripotency; they have a stable, normal karyotype in vitro; they can be propagated indefinitely in theory without differentiation; they can differentiate spontaneously into multiple cell types representative of all three embryonic germ layers, both in teratomas after grafting and in vitro under appropriate conditions; and they can give rise to any cell type in the body, including germ cells, when injected into host blastocysts[12]. Many researchers attempted to establish rat ES cells by using the same conditions as those of mouse ES cells, namely leukemia inhibitory factor (LIF) in combination with bone morphogenetic protein (BMP) or fetal bovine serum (FBS). The rat blastocyst-derived cells cultured under these conditions expressed various embryonic stem cell specific markers such as stage-specific embryonic antigen-I (SSEA-1), Oct4, and alkaline phosphatase, whereas the chimeric rats or teratocarcinoma derived from the rat blastocyst-derived cells were not confirmed[13-15].

Figure 1
Figure 1 Establishment of rat embryonic stem cells. A: E4.5 Blastocysts are isolated from Sprague-Dawley rats, and the blastocysts dissolved zona pellucida are put on mouse embryonic fibroblasts (MEF) feeders; B: After 7-10 d, the outgrowth formed from the blastocysts are dispersed, and transferred on MEF feeders; C: Approximately 7 d after culture, rat embryonic stem (ES) cells are appeared (our unpublished data).
Establishment of rat embryonic stem cells using small molecules

In 2008, a robust and efficient method to establish mouse ES cells was reported by Ying et al[16], which are also applicable to establish rat ES cells. To maintain the pluripotent state of mouse ES cells, LIF activates the Janus kinase/signal transducer and activator of transcription signaling pathway, while BMP and serum activate the Sma and Mad Related Family signaling pathway[17]. Differentiation of mouse ES cells is induced via activation of mitogen-activated protein kinase/extracellular signal-related kinase (ERK) kinase (MEK) pathway by the autocrine stimulation of fibroblast growth factor-4 (FGF4)[18]. Ying et al[16] revealed that LIF, serum and BMP affect the downstream of ERK, and demonstrated that the inhibition of MEK and the FGF receptor (FGFR) maintains the pluripotency of mouse ES cells under serum-free conditions. Moreover, the self-renewal capacity of mouse ES cells was promoted by an additional inhibition of glycogen synthase kinase 3 (GSK3), as the GSK3 pathway is involved in the maintenance of ES cells in the undifferentiated state viaβ-catenin/Wnt signaling[19-25]. These findings revealed that two types of culture conditions, a combination of the MEK inhibitor and GSK3 inhibitor (2i condition), and a combination of the MEK inhibitor, FGFR inhibitor, and GSK3 inhibitor (3i condition), enable not only the maintenance of mouse ES cells but also the “robust” and “effective” establishment of rat ES cells displaying all the defining properties of ES cells[9,10,26-31]. The morphology of rat ES cells is rounded and loosely attached on feeder cells, unlike mouse ES cells; however, the rat ES cells express the same undifferentiated markers as mouse ES cells: Oct4, Nanog, Sox2, Rex1, FGF4, and SSEA1. Kawamata et al[26] succeeded in the establishment of rat ES cells even in FBS-containing medium using Rho-associated, coiled-coil containing protein kinase (ROCK) inhibitor and activin receptor-like kinase (ALK5) inhibitor in addition to the MEK inhibitor and GSK3 inhibitor, which cells showed the defining properties such as pluripotent markers, embryonic body formation, and normal karyotype (Figure 2; our unpublished data). Table 1 shows various attempts to establish rat ES cells, and the properties of the resultant cells.

Table 1 Summary of components for the establishment of rat embryonic stem cells and the properties of the resultant cells.
Ref.AdditivesPluripotent markersKaryotype stabilityEB/teratocarcinomaGermline transmission
Iannacconne et al[13]FBSAPUnconfirmedFailedFailed
LIFSSEA1
Takahama et al[14]FBSAPUnconfirmedUnconfirmedUnconfirmed
LIFOct3/4
SSEA1
Vassilieva et al[15]FBSAPUnconfirmedUnconfirmedUnconfirmed
LIFOct3/4
SSEA1
Buehr et al[9](A) N2B27ConfirmedConfirmedConfirmed
FGFRiAP
MEKiOct3/4
GSK3iSSEA1
(B)Sox2
N2B27
MEKiNanog
GSK3iFgf4
LIFRex1
Li et al[10](A) N2B27ConfirmedConfirmedConfirmed
MEKiAP
GSK3iOct3/4
(B)SSEA1
N2B27
MEKiSox2
GSK3iNanog
LIF
Kawamata et al[26]FBSAPConfirmedConfirmedConfirmed
MEKiOct3/4
GSK3iSox2
ALK5iNanog
ROCKiRex1
Figure 2
Figure 2 Properties of the rat embryonic stem cells. The rat embryonic stem cells are established using the combination of four inhibitors and serum: mitogen-associated protein kinase/extracellular signal-related kinase kinase (MEK) inhibitor, glycogen synthase kinase 3 inhibitor, activin receptor-like kinase (ALK5) inhibitor, Rho-associated, coiled-coil containing protein kinase inhibitor and FBS. A: Expression of the pluripotent markers Oct4 and Sox2, and alkaline phosphatase staining; B: Embryonic body formation; C: Karyotype analysis by G-band staining (our unpublished data). DAPI: 4',6 -diamidino-2-phenylindole; FBS: Fetal bovine serum.
Improvement of rat embryonic stem cell culture efficiency

Numerous approaches have been made to efficiently propagate rat ES cells. Soon after the establishment of rat ES cells[9,10], rat induced pluripotent stem (iPS) cells were established using the 2i culture conditions[32,33]. The additional inhibition of ALK5 allowed to propagate rat iPS cells as a homogeneous population, with less spontaneously differentiated colonies as compared to the 2i condition without ALK5 inhibition[32]. Furthermore, the inhibition of ROCK prevents from apoptosis in human ES/iPS cells and enhances their attachment on feeder cells after enzymatic dissociation[34,35]. These studies suggested that the addition of the ALK5 inhibitor and ROCK inhibitor to either the 2i or 3i condition promotes the efficient culture of rat ES cells. Kawamata et al[26] confirmed the effect of the ROCK inhibitor on the self-renewal of rat ES cells by using Oct4-Venous-transgenic rat ES cells, in which cells were derived from the transgenic rats expressing the fluorescent Venus protein under the Oct4 promoter/enhancer. In a condition containing all the four inhibitors (the ROCK inhibitor Y-27632, the MEK inhibitor PD0325901, the ALK5 inhibitor A-83-01, and the GSK3 inhibitor CHIR99021) referred to as YPAC, a great number of colonies expressing the pluripotent markers Oct4 and alkaline phosphatase were appeared after single-cell enzymatic dissociation. In contrast, the rat ES cell colonies maintained withdrawal of the ROCK inhibitor from the YPAC condition expressed the pluripotent markers, but the colonies were sparse under the three-inhibitor condition compared to the YPAC condition[26]. Kawamata et al[26] concluded that the ROCK inhibitor promotes the attachment of rat ES cells on feeder cells, which leads to efficient culture expansion. Li et al[36] compared the 2i condition with the YPAC condition, and concluded that rat ES cells could expand approximately twice more under the YPAC condition than under the 2i condition, while maintaining the undifferentiated state. A combination of the serum and the 2i condition was also attempted for the culture of rat ES cells, and as a result, the addition of the serum enhanced the attachment of rat ES cells to feeder cells[29]. However, the addition of serum triggers differentiation in the cultured ES cells, and may introduce unexpected side effects due to unknown factors within the serum[37]. Actually, the expression level of a trophectoderm marker Cdx2 was three times higher in the rat ES cells cultured in the serum with YPAC than in the cells cultured in the serum-free 2i condition[38].

Regulation of Cdx2 expression in rat embryonic stem cells

Cdx2 is the marker of trophectoderm cells, which is not expressed in mouse ES cells; hence, some studies attempted to find culture condition to repress the expression of Cdx2[39,40]. It was reported that the expression of Cdx2 was related to the concentration of the GSK3 inhibitor. The low concentration of the GSK3 inhibitor 1.5 μmol/L was found to maintain pluripotency and reduce Cdx2 expression; however, the higher concentration of the GSK3 inhibitor 3 μmol/L, which has generally been used to establish and maintain rat ES cells, promotes the expression of Cdx2[41]. In addition, a decrease in Cdx2 expression was reported by maintaining rat ES cells on Matrigel® or in suspension[29].

Maintenance of chromosomal states in rat embryonic stem cells

One of the defining properties of ES cells is the retention of a normal karyotype after prolonged culture. However, the rat ES cells cultured under the 2i condition retained normal karyotypes fewer than 40%, which were lower than that found in mouse ES cells[42]. It is known that the low germline-competence of mouse ES cells results from abnormal karyotypes[43], and hence, the instability of karyotypes in rat ES cells would make germline transmission difficult. It is reported that the combination of the serum and YPAC improves the karyotype stability, which retains normal karyotypes with over 70%[26]. The effect of YPAC on karyotype stability was clarified by Li et al[36] by the successful generation of germline-competent chimeric rats by using the highly passaged rat ES cells maintained under the serum-free YPAC condition, while the rat ES cells maintained under the 2i condition failed to contribute germline. These results suggest that the inhibition of ROCK and/or ALK5 in addition to the 2i condition can stabilize the karyotypes of rat ES cells.

DIFFERENTIATION OF RAT EMBRYONIC STEM CELLS INTO FUNCTIONAL CELLS

Only a few studies have reported a stable induction method to differentiate rat ES cells into functional cells in vitro. Cao et al[44] supplied a protocol to differentiate rat ES cells into cardiomyocytes, and showed that the functional properties of the differentiated cells were similar to those of rat fetal cardiomyocytes and mouse ES cell-derived cardiomyocytes. However, the efficiency of the cardiac differentiation of rat ES cells was approximately 40%; this was lower than that of mouse ES cells, which were differentiated with over 80% efficiency[45]. Normally, the first step for in vitro differentiation of mouse ES cells is to induce cell aggregation into embryonic bodies (EBs), which can be accomplished in a relatively simple procedure using a differentiation medium containing serum. However, apoptosis was observed in rat ES cells during the formation of EBs in the differentiation medium[26,44]. On the other hand, Peng et al[46] succeeded in efficient differentiation of neural precursors from rat ES cells. They demonstrated that the ROCK inhibitor facilitates the neural differentiation and the GSK3 inhibitor maintains the survivability of the differentiated cells, and high-efficiency neural precursor differentiation (90%) was achieved using the two inhibitors. For the survival of neural precursors differentiated from mouse ES cells, it is sufficient only in the ROCK inhibitor[47]. Thus, optimized differentiation media for rat ES cells are necessary to induce rat ES cells into functional cells efficiently. These media may facilitate not only in vitro studies but also in vivo studies such as the transplantation of rat ES cell-derived differentiated cells or grafts to animal models, which in vivo studies were not reported even though rats are more relevant to humans than mice.

GENERATION OF KNOCK-IN AND KNOCK-OUT RATS USING RAT EMBRYONIC STEM CELLS
Gene-targeting to rat embryonic stem cells

Gene targeting to rat ES cells has been explored in some previous studies. Tong et al[11] and Yamamoto et al[48] reported that the efficiency of homologous recombination was 1.12%-3.70% and 0.9%, respectively, while Meek et al[28] reported that the efficiency was similar to that of mouse and human ES cells. Moreover, the gene-targeted rat ES cells express the pluripotent markers to the same extent as the parent ES cells, and retain the ability to differentiate into all three germ layers[28]. It was noted that serum was effective at minimizing the damage caused when electroporation was performed[49]. In fact, Yamamoto et al[48] used 1% serum during their electroporation procedure. Such efforts are not necessary for the rat ES cells maintained in the condition developed by Kawamata et al[26], because the culture condition already contains 20% FBS.

Embryonic stem cell maintenance for creating
knock-out rats

Soon after the generation of transgenic rats using rat ES cells[26,50], the first study of targeted gene disruption in rats via homologous recombination in rat ES cells were reported by Tong et al[11]. Figure 3 shows a process of generating genetically-modified rats. Tong et al[11] succeeded in generating germline chimeric rats by selecting the colonies that formed a small, round, compact shape, and loose attachment on feeder cells, not the large or strongly adherent colonies. This finding provides a guideline to choose rat ES cell colonies for microinjection. Tong et al[11] found that over 65% of the large and strongly adherent colonies were polyploid, and concluded that such chromosomal abnormalities caused the failure of germline transmission, as the same as the case in mouse ES cells. Hence, the likelihood of successful germline transmission could be increased by the selection of small, loosely adherent rat ES cell colonies for the purpose of microinjection.

Figure 3
Figure 3 Generation of embryonic stem cell-derived gene-modified rats. First, a gene of interest is introduced into rat embryonic stem (ES) cells. Next, the gene-targeted rat ES cells are injected into blastocysts and are incubated in embryo culture medium or YPAC medium. Then, the chimeric rat embryos are transferred into recipient rats. After that, gene-modified rats will be generated following breeding of the chimeric rats obtained from chimeric embryos.

An unstable state of rat ES cells in recipient blastocysts after microinjection was also considered as a possible cause for failure to produce germline-competent chimeric rats. In order to study the fate of rat ES cells in recipient blastocysts, Kawamata et al[26] delivered CAG-AmCyan1-transgenic rat ES cells into recipient blastocysts by microinjection. In the absence of YPAC, few cyan-positive cells were present in the blastocysts after 30 h incubation, whereas cyan-positive rat ES cells remained on the surface of the ICM when YPAC was used in both the injection and incubation media. It is reasonable that the addition of the ROCK inhibitor not only suppresses apoptosis, but also promotes adherence to the ICM. Some embryo culture media such as M2, M16, and KSOM have been generally used for the microinjection of mouse ES cells. Similarly, the use of the embryo culture media is also proposed for microinjection of rat ES cells[51]. Specifically, embryos are washed in M2 medium after collection from pregnant rats, and the chimeric embryos are incubated in M16 medium after microinjection to allow recovery. However, the use of the YPAC medium might offer a superior microinjection method to the conventional methods.

Embryonic stem cell-based knock-out rats and their phenotypes

Two kinds of gene-targeted rats - p53-knock-out rats and protease-activated receptor-2 (Par-2) knock-out rats - have been developed using rat ES cell technology, by three groups[11,48,52]. The p53-knock-out rats and p53-knock-out mice revealed differing phenotypes: only 4% of female rats homozygous for the p53 knock-out could survive, whereas the female mice homozygous for the p53 knock-out were normally present at weaning[52]; the major tumor type of the p53-knock-out rats is hemangiosarcoma, while that of the p53-knock-out mice is lymphoma[53]; and developmental abnormalities associated with neural tube defects occurred more frequently in the female p53-knock-out rats than in those female mice[54]. Such sexual distortion associated with the neural tube defects also cause in human[55,56].

A phenotype of Par-2-knock-out rats were reported by Yamamoto et al[48]. The Par-2-knock-out rats showed a lack of responsiveness to PAR-2 receptor activating peptides, which clearly indicated the deficiency of Par-2 protein.

Rat models in studying human diseases

The transgenic rat model of Huntington’s disease reflects more typical adult patient pathologies in comparison to the transgenic mice[57]. The transgenic rat model of Alzheimer’s disease, harboring mutant human transgenes for amyloid precursor protein and presenilin 1, manifests a complete repertoire of Alzheimer’s disease pathological features and demonstrates a markedly greater abundance of soluble oligomeric amyloid-β peptides than mice harboring the same human transgenes[58]. Moreover, the usefulness of transgenic rat models was shown in autism and fragile X syndrome researches. Transgenic rat models lacking disease-associated genes such as fragile X mental retardation 1 gene and neuroligin-3, and these mutations display traits that may be analogous to the characteristics of in their respective human diseases[59].

Although gene knock-out strategies are a promising tool to clarify the mechanism of human diseases such as those mentioned above, some human disease-related gene knock-outs are developmentally lethal. This problem can often be overcome through the use of conditional-knock-out animals, which enable site-specific and inducible gene deletion. Brown et al[60] first generated the conditional-knock-out rats via ZFN-mediated technologies. The system is based on Cre/loxP recombination system, and a target gene is modified by the insertion of two flanking loxP sites, enabling excision of the flanked (floxed) gene segment through Cre-mediated recombination. Conditional-knock-out mice are obtained by mating the floxed mouse line with a Cre-transgenic line, such that the target gene in the progeny becomes inactivated in vivo within the expression domain of Cre[61,62]. To create the conditional-knock-out rats, Brown et al[60] mated the two strains: floxed allele-harboring rats and Cre allele-harboring rats, generated via pronuclear co-injection of a pair of ZFNs along with each donor plasmid.

PERSPECTIVES

Rats are used as the first-preferred animal species for in vivo tests of chemicals. In the meantime, in vitro alternative methods reflecting a response to chemicals have been demanded due to concerns about animal welfare. The use of the in vitro alternatives has many advantages: a decrease in the number of animals used, a reduced cost of animal maintenance, a smaller quantity of chemical needed for testing, and a shortening of the time essential to prepare for and conduct experiments[63-65]. Rat ES cells could provide innovative in vitro screening models, as rat ES cells have an infinite proliferative capacity and can be continuously supplied.

The generation of transgenic rats have been supported by not only rat ES cell-based gene-modification technology but also other new tools for gene modification such as ZFNs, transcription activator-like effector nuclease and clustered regularly interspaced short palindromic repeat[66-68]. Table 2 shows the knock-out rats generated by using of rat ES cells and new gene-modification technologies. However, the new technologies carry a risk of off-target effects by site-specific nuclease activity, and require screening to detect targeted events. In contrast, using ES cell-based technologies allows researchers to easily approach the generation of genetically modified rats with the knowledge obtained from experience in generating genetically modified mice. Therefore, rat ES cell-based gene targeting is still an essential tool for gene modifications.

Table 2 List of knock-out rats generated using new gene-targeted technologies and rat embryonic stem cell-based technologies.
Ref.Cell typeTechnologyTarget
Ménoret et al[69]EmbryoMeganucleaseRag1
Ménoret et al[70]EmbryoZFNImmunoglobulin heavy chain
Zschemisch et al[71]EmbryoZFNRag1
Geurts et al[7]EmbryoZFNGFP
EmbryoZFNIgM
EmbryoZFNRab38
Mashimo et al[8]EmbryoZFNIL-2 receptor gamma
Moreno et al[72]EmbryoZFNRenin
Chu et al[73]EmbryoZFNMdr1
Vaira et al[74]EmbryoZFNReptin
Tesson et al[66]EmbryoTALENIgM
Ferguson et al[67]EmbryoTALENTlr4
Yamamoto et al[48]ES cellsHomologous RecombinationPar2
Tong et al[11]ES cellsHomologous Recombinationp53
Kawamata et al[52]ES cellsZFNp53
CONCLUSION

Exploring small molecules for the optimum culture conditions for rat ES cells opened up avenues for the generation of genetically modified rats. Although ES cell-based knock-out/knock-in rats have not been widely generated due to the karyotype instability of rat ES cells, further exploration of new combinations of small molecules and growth factors will facilitate germline transmission of genetically modified rat ES cell clones. Additionally, human ES cells with a naïve state could be created by above effort[75]. As the phenotypes of gene-knock-out animals can be different among species, rats represent a valuable tool in which phenotypes can be generated and compared to those of mice with relative ease. Studies using a combination of both mouse and rat disease models will provide beneficial information to clarify the mechanisms of human disease, leading to the development of new drugs. We hope that many researchers again choose to utilize rat models for research, and establish a new platform for basic and clinical applications using gene modification technology.

ACKNOWLEDGMENTS

The authors would like to express the deepest appreciation to Yoshihiro Sumita and Tadayoshi Ueda whose comments and suggestions were of inestimable value for this manuscript. The authors also thanks to Aya Yoshimura (National Institute of Neuroscience) for technical advice, Luc Gailhouste (National Cancer Center Research Institute) for comments on this manuscript and Shigeyuki Ota, Lao Xintian, Makiko Morita, Hiroko Koreishi, Kenryo Furushima and Yasuyuki Tsutsumishita for their sincere encouragement.

Footnotes

P- Reviewer: Feng ZL, Sidhu KS S- Editor: Gong XM L- Editor: A E- Editor: Wu HL

References
1.  Voigt B, Serikawa T. Pluripotent stem cells and other technologies will eventually open the door for straightforward gene targeting in the rat. Dis Model Mech. 2009;2:341-343.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 14]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
2.  Hedrich JH. History, Strains and Models. The Laboratory Rat. 2000;3-16.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Jacob HJ, Kwitek AE. Rat genetics: attaching physiology and pharmacology to the genome. Nat Rev Genet. 2002;3:33-42.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 185]  [Cited by in F6Publishing: 200]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
4.  Abbott A. Laboratory animals: the Renaissance rat. Nature. 2004;428:464-466.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 115]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
5.  Zhao S, Liu E, Chu Y, Zheng H, Kitajima S, Morimoto M. Numbers of Publications Related to Laboratory Animals. Scand J Lab Anim Sci. 2007;34:81-86.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Evans MJ, Kaufman MH. Establishment in culture of pluripotential cells from mouse embryos. Nature. 1981;292:154-156.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5956]  [Cited by in F6Publishing: 5274]  [Article Influence: 122.7]  [Reference Citation Analysis (0)]
7.  Geurts AM, Cost GJ, Freyvert Y, Zeitler B, Miller JC, Choi VM, Jenkins SS, Wood A, Cui X, Meng X. Knockout rats via embryo microinjection of zinc-finger nucleases. Science. 2009;325:433.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 712]  [Cited by in F6Publishing: 689]  [Article Influence: 45.9]  [Reference Citation Analysis (0)]
8.  Mashimo T, Takizawa A, Voigt B, Yoshimi K, Hiai H, Kuramoto T, Serikawa T. Generation of knockout rats with X-linked severe combined immunodeficiency (X-SCID) using zinc-finger nucleases. PLoS One. 2010;5:e8870.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 192]  [Cited by in F6Publishing: 200]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
9.  Buehr M, Meek S, Blair K, Yang J, Ure J, Silva J, McLay R, Hall J, Ying QL, Smith A. Capture of authentic embryonic stem cells from rat blastocysts. Cell. 2008;135:1287-1298.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 611]  [Cited by in F6Publishing: 492]  [Article Influence: 32.8]  [Reference Citation Analysis (0)]
10.  Li P, Tong C, Mehrian-Shai R, Jia L, Wu N, Yan Y, Maxson RE, Schulze EN, Song H, Hsieh CL. Germline competent embryonic stem cells derived from rat blastocysts. Cell. 2008;135:1299-1310.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 522]  [Cited by in F6Publishing: 499]  [Article Influence: 33.3]  [Reference Citation Analysis (0)]
11.  Tong C, Li P, Wu NL, Yan Y, Ying QL. Production of p53 gene knockout rats by homologous recombination in embryonic stem cells. Nature. 2010;467:211-213.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 220]  [Cited by in F6Publishing: 209]  [Article Influence: 14.9]  [Reference Citation Analysis (0)]
12.  Pera MF, Reubinoff B, Trounson A. Human embryonic stem cells. J Cell Sci. 2000;113:5-10.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Iannaccone PM; Rat pluripotent embryonic stem cells and method of obtaining and using same. International Application No.: PCT/US1994/009787.  .  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Takahama Y, Ochiya T, Sasaki H, Baba-Toriyama H, Konishi H, Nakano H, Terada M. Molecular cloning and functional analysis of cDNA encoding a rat leukemia inhibitory factor: towards generation of pluripotent rat embryonic stem cells. Oncogene. 1998;16:3189-3196.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
15.  Vassilieva S, Guan K, Pich U, Wobus AM. Establishment of SSEA-1- and Oct-4-expressing rat embryonic stem-like cell lines and effects of cytokines of the IL-6 family on clonal growth. Exp Cell Res. 2000;258:361-373.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 89]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
16.  Ying QL, Wray J, Nichols J, Batlle-Morera L, Doble B, Woodgett J, Cohen P, Smith A. The ground state of embryonic stem cell self-renewal. Nature. 2008;453:519-523.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2607]  [Cited by in F6Publishing: 2548]  [Article Influence: 159.3]  [Reference Citation Analysis (0)]
17.  Ying QL, Nichols J, Chambers I, Smith A. BMP induction of Id proteins suppresses differentiation and sustains embryonic stem cell self-renewal in collaboration with STAT3. Cell. 2003;115:281-292.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1593]  [Cited by in F6Publishing: 1517]  [Article Influence: 75.9]  [Reference Citation Analysis (0)]
18.  Kunath T, Saba-El-Leil MK, Almousailleakh M, Wray J, Meloche S, Smith A. FGF stimulation of the Erk1/2 signalling cascade triggers transition of pluripotent embryonic stem cells from self-renewal to lineage commitment. Development. 2007;134:2895-2902.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 591]  [Cited by in F6Publishing: 616]  [Article Influence: 36.2]  [Reference Citation Analysis (0)]
19.  Doble BW, Woodgett JR. GSK-3: tricks of the trade for a multi-tasking kinase. J Cell Sci. 2003;116:1175-1186.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1584]  [Cited by in F6Publishing: 1622]  [Article Influence: 77.2]  [Reference Citation Analysis (0)]
20.  Jope RS, Johnson GV. The glamour and gloom of glycogen synthase kinase-3. Trends Biochem Sci. 2004;29:95-102.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1141]  [Cited by in F6Publishing: 1192]  [Article Influence: 59.6]  [Reference Citation Analysis (0)]
21.  Sato N, Meijer L, Skaltsounis L, Greengard P, Brivanlou AH. Maintenance of pluripotency in human and mouse embryonic stem cells through activation of Wnt signaling by a pharmacological GSK-3-specific inhibitor. Nat Med. 2004;10:55-63.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1567]  [Cited by in F6Publishing: 1537]  [Article Influence: 73.2]  [Reference Citation Analysis (0)]
22.  Wu D, Pan W. GSK3: a multifaceted kinase in Wnt signaling. Trends Biochem Sci. 2010;35:161-168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 559]  [Cited by in F6Publishing: 614]  [Article Influence: 40.9]  [Reference Citation Analysis (0)]
23.  Lyashenko N, Winter M, Migliorini D, Biechele T, Moon RT, Hartmann C. Differential requirement for the dual functions of β-catenin in embryonic stem cell self-renewal and germ layer formation. Nat Cell Biol. 2011;13:753-761.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 188]  [Cited by in F6Publishing: 196]  [Article Influence: 15.1]  [Reference Citation Analysis (0)]
24.  Wray J, Kalkan T, Gomez-Lopez S, Eckardt D, Cook A, Kemler R, Smith A. Inhibition of glycogen synthase kinase-3 alleviates Tcf3 repression of the pluripotency network and increases embryonic stem cell resistance to differentiation. Nat Cell Biol. 2011;13:838-845.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 428]  [Cited by in F6Publishing: 395]  [Article Influence: 30.4]  [Reference Citation Analysis (0)]
25.  Martello G, Sugimoto T, Diamanti E, Joshi A, Hannah R, Ohtsuka S, Göttgens B, Niwa H, Smith A. Esrrb is a pivotal target of the Gsk3/Tcf3 axis regulating embryonic stem cell self-renewal. Cell Stem Cell. 2012;11:491-504.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 326]  [Cited by in F6Publishing: 331]  [Article Influence: 27.6]  [Reference Citation Analysis (0)]
26.  Kawamata M, Ochiya T. Generation of genetically modified rats from embryonic stem cells. Proc Natl Acad Sci USA. 2010;107:14223-14228.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 88]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
27.  Hirabayashi M, Kato M, Kobayashi T, Sanbo M, Yagi T, Hochi S, Nakauchi H. Establishment of rat embryonic stem cell lines that can participate in germline chimerae at high efficiency. Mol Reprod Dev. 2010;77:94.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 35]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
28.  Meek S, Buehr M, Sutherland L, Thomson A, Mullins JJ, Smith AJ, Burdon T. Efficient gene targeting by homologous recombination in rat embryonic stem cells. PLoS One. 2010;5:e14225.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 45]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
29.  Hong J, He H, Weiss ML. Derivation and characterization of embryonic stem cells lines derived from transgenic Fischer 344 and Dark Agouti rats. Stem Cells Dev. 2012;21:1571-1586.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
30.  Men H, Bauer BA, Bryda EC. Germline transmission of a novel rat embryonic stem cell line derived from transgenic rats. Stem Cells Dev. 2012;21:2606-2612.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 16]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
31.  Men H, Bryda EC. Derivation of a germline competent transgenic Fischer 344 embryonic stem cell line. PLoS One. 2013;8:e56518.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 16]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
32.  Li W, Wei W, Zhu S, Zhu J, Shi Y, Lin T, Hao E, Hayek A, Deng H, Ding S. Generation of rat and human induced pluripotent stem cells by combining genetic reprogramming and chemical inhibitors. Cell Stem Cell. 2009;4:16-19.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 432]  [Cited by in F6Publishing: 403]  [Article Influence: 25.2]  [Reference Citation Analysis (0)]
33.  Hamanaka S, Yamaguchi T, Kobayashi T, Kato-Itoh M, Yamazaki S, Sato H, Umino A, Wakiyama Y, Arai M, Sanbo M. Generation of germline-competent rat induced pluripotent stem cells. PLoS One. 2011;6:e22008.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 54]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
34.  Ishizaki T, Uehata M, Tamechika I, Keel J, Nonomura K, Maekawa M, Narumiya S. Pharmacological properties of Y-27632, a specific inhibitor of rho-associated kinases. Mol Pharmacol. 2000;57:976-983.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Watanabe K, Ueno M, Kamiya D, Nishiyama A, Matsumura M, Wataya T, Takahashi JB, Nishikawa S, Nishikawa S, Muguruma K. A ROCK inhibitor permits survival of dissociated human embryonic stem cells. Nat Biotechnol. 2007;25:681-686.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1505]  [Cited by in F6Publishing: 1496]  [Article Influence: 88.0]  [Reference Citation Analysis (0)]
36.  Li T, Zhao XY, Teng F, Li X, Jiang M, Li W, Wang X, Wang J, Liu L, Liu Z. Derivation of germline competent rat embryonic stem cells from DA rats. J Genet Genomics. 2012;39:603-606.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
37.  Tong C, Huang G, Ashton C, Li P, Ying QL. Generating gene knockout rats by homologous recombination in embryonic stem cells. Nat Protoc. 2011;6:827-844.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 67]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
38.  Hong J, He H, Bui P, Ryba-White B, Rumi MA, Soares MJ, Dutta D, Paul S, Kawamata M, Ochiya T. A focused microarray for screening rat embryonic stem cell lines. Stem Cells Dev. 2013;22:431-443.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
39.  Ralston A, Rossant J. Genetic regulation of stem cell origins in the mouse embryo. Clin Genet. 2005;68:106-112.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 132]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
40.  Sasaki H. Mechanisms of trophectoderm fate specification in preimplantation mouse development. Dev Growth Differ. 2010;52:263-273.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 67]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
41.  Chen Y, Blair K, Smith A. Robust self-renewal of rat embryonic stem cells requires fine-tuning of glycogen synthase kinase-3 inhibition. Stem Cell Reports. 2013;1:209-217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 51]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
42.  Blair K, Leitch HG, Mansfield W, Dumeau CÉ, Humphreys P, Smith AG. Culture parameters for stable expansion, genetic modification and germline transmission of rat pluripotent stem cells. Biol Open. 2012;1:58-65.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 28]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
43.  Liu X, Wu H, Loring J, Hormuzdi S, Disteche CM, Bornstein P, Jaenisch R. Trisomy eight in ES cells is a common potential problem in gene targeting and interferes with germ line transmission. Dev Dyn. 1997;209:85-91.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Cao N, Liao J, Liu Z, Zhu W, Wang J, Liu L, Yu L, Xu P, Cui C, Xiao L. In vitro differentiation of rat embryonic stem cells into functional cardiomyocytes. Cell Res. 2011;21:1316-1331.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 31]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
45.  Boheler KR, Czyz J, Tweedie D, Yang HT, Anisimov SV, Wobus AM. Differentiation of pluripotent embryonic stem cells into cardiomyocytes. Circ Res. 2002;91:189-201.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 541]  [Cited by in F6Publishing: 503]  [Article Influence: 22.9]  [Reference Citation Analysis (0)]
46.  Peng X, Gao H, Wang Y, Yang B, Liu T, Sun Y, Jin H, Jiang L, Li L, Wu M. Conversion of rat embryonic stem cells into neural precursors in chemical-defined medium. Biochem Biophys Res Commun. 2013;431:783-787.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 7]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
47.  Koyanagi M, Takahashi J, Arakawa Y, Doi D, Fukuda H, Hayashi H, Narumiya S, Hashimoto N. Inhibition of the Rho/ROCK pathway reduces apoptosis during transplantation of embryonic stem cell-derived neural precursors. J Neurosci Res. 2008;86:270-280.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 129]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
48.  Yamamoto S, Nakata M, Sasada R, Ooshima Y, Yano T, Shinozawa T, Tsukimi Y, Takeyama M, Matsumoto Y, Hashimoto T. Derivation of rat embryonic stem cells and generation of protease-activated receptor-2 knockout rats. Transgenic Res. 2012;21:743-755.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 25]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
49.  Bahnson AB, Boggs SS. Addition of serum to electroporated cells enhances survival and transfection efficiency. Biochem Biophys Res Commun. 1990;171:752-757.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
50.  Hirabayashi M, Kato M, Sanbo M, Kobayashi T, Hochi S, Nakauchi H. Rat transgenesis via embryonic stem cells electroporated with the Kusabira-orange gene. Mol Reprod Dev. 2010;77:474.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 29]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
51.  Li P, Schulze EN, Tong C, Ying QL.  Rat embryonic stem cell derivation and propagation. Advanced protocols for animal transgenesis (Springer Protocols Handbooks). Berlin: Springer 2011; 457-475.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Kawamata M, Ochiya T. Two distinct knockout approaches highlight a critical role for p53 in rat development. Sci Rep. 2012;2:945.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 14]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
53.  Huang G, Tong C, Kumbhani DS, Ashton C, Yan H, Ying QL. Beyond knockout rats: new insights into finer genome manipulation in rats. Cell Cycle. 2011;10:1059-1066.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 27]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
54.  Armstrong JF, Kaufman MH, Harrison DJ, Clarke AR. High-frequency developmental abnormalities in p53-deficient mice. Curr Biol. 1995;5:931-936.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 338]  [Cited by in F6Publishing: 358]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
55.  Copp AJ, Brook FA, Estibeiro JP, Shum AS, Cockroft DL. The embryonic development of mammalian neural tube defects. Prog Neurobiol. 1990;35:363-403.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 236]  [Cited by in F6Publishing: 220]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
56.  Chen X, Watkins R, Delot E, Reliene R, Schiestl RH, Burgoyne PS, Arnold AP. Sex difference in neural tube defects in p53-null mice is caused by differences in the complement of X not Y genes. Dev Neurobiol. 2008;68:265-273.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 72]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
57.  von Hörsten S, Schmitt I, Nguyen HP, Holzmann C, Schmidt T, Walther T, Bader M, Pabst R, Kobbe P, Krotova J. Transgenic rat model of Huntington’s disease. Hum Mol Genet. 2003;12:617-624.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 219]  [Cited by in F6Publishing: 209]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
58.  Cohen RM, Rezai-Zadeh K, Weitz TM, Rentsendorj A, Gate D, Spivak I, Bholat Y, Vasilevko V, Glabe CG, Breunig JJ. A transgenic Alzheimer rat with plaques, tau pathology, behavioral impairment, oligomeric aβ, and frank neuronal loss. J Neurosci. 2013;33:6245-6256.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 256]  [Cited by in F6Publishing: 295]  [Article Influence: 26.8]  [Reference Citation Analysis (0)]
59.  Hamilton SM, Green JR, Veeraragavan S, Yuva L, McCoy A, Wu Y, Warren J, Little L, Ji D, Cui X. Fmr1 and Nlgn3 knockout rats: novel tools for investigating autism spectrum disorders. Behav Neurosci. 2014;128:103-109.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 102]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
60.  Brown AJ, Fisher DA, Kouranova E, McCoy A, Forbes K, Wu Y, Henry R, Ji D, Chambers A, Warren J. Whole-rat conditional gene knockout via genome editing. Nat Methods. 2013;10:638-640.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 67]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
61.  Friedel RH, Wurst W, Wefers B, Kühn R. Generating conditional knockout mice. Methods Mol Biol. 2011;693:205-231.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 56]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
62.  Sauer B. Functional expression of the cre-lox site-specific recombination system in the yeast Saccharomyces cerevisiae. Mol Cell Biol. 1987;7:2087-2096.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 278]  [Cited by in F6Publishing: 251]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
63.  DelRaso NJ. In vitro methodologies for enhanced toxicity testing. Toxicol Lett. 1993;68:91-99.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 25]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
64.  LeCluyse EL, Bullock PL, Parkinson A. Strategies for restoration and maintenance of normal hepatic structure and function in long-term cultures of rat hepatocytes. Adv Drug Deliver Rev. 1996;22:133-186.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 143]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
65.  Soldatow VY, Lecluyse EL, Griffith LG, Rusyn I. In vitro models for liver toxicity testing. Toxicol Res (Camb). 2013;2:23-39.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 300]  [Cited by in F6Publishing: 305]  [Article Influence: 25.4]  [Reference Citation Analysis (0)]
66.  Tesson L, Usal C, Ménoret S, Leung E, Niles BJ, Remy S, Santiago Y, Vincent AI, Meng X, Zhang L. Knockout rats generated by embryo microinjection of TALENs. Nat Biotechnol. 2011;29:695-696.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 473]  [Cited by in F6Publishing: 492]  [Article Influence: 37.8]  [Reference Citation Analysis (0)]
67.  Ferguson C, McKay M, Harris RA, Homanics GE. Toll-like receptor 4 (Tlr4) knockout rats produced by transcriptional activator-like effector nuclease (TALEN)-mediated gene inactivation. Alcohol. 2013;47:595-599.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 28]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
68.  Shan Q, Wang Y, Li J, Zhang Y, Chen K, Liang Z, Zhang K, Liu J, Xi JJ, Qiu JL. Targeted genome modification of crop plants using a CRISPR-Cas system. Nat Biotechnol. 2013;31:686-688.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1249]  [Cited by in F6Publishing: 1084]  [Article Influence: 108.4]  [Reference Citation Analysis (0)]
69.  Ménoret S, Fontanière S, Jantz D, Tesson L, Thinard R, Rémy S, Usal C, Ouisse LH, Fraichard A, Anegon I. Generation of Rag1-knockout immunodeficient rats and mice using engineered meganucleases. FASEB J. 2013;27:703-711.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 79]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
70.  Ménoret S, Iscache AL, Tesson L, Rémy S, Usal C, Osborn MJ, Cost GJ, Brüggemann M, Buelow R, Anegon I. Characterization of immunoglobulin heavy chain knockout rats. Eur J Immunol. 2010;40:2932-2941.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 53]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
71.  Zschemisch NH, Glage S, Wedekind D, Weinstein EJ, Cui X, Dorsch M, Hedrich HJ. Zinc-finger nuclease mediated disruption of Rag1 in the LEW/Ztm rat. BMC Immunol. 2012;13:60.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 28]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
72.  Moreno C, Hoffman M, Stodola TJ, Didier DN, Lazar J, Geurts AM, North PE, Jacob HJ, Greene AS. Creation and characterization of a renin knockout rat. Hypertension. 2011;57:614-619.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 63]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
73.  Chu X, Zhang Z, Yabut J, Horwitz S, Levorse J, Li XQ, Zhu L, Lederman H, Ortiga R, Strauss J. Characterization of multidrug resistance 1a/P-glycoprotein knockout rats generated by zinc finger nucleases. Mol Pharmacol. 2012;81:220-227.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 43]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
74.  Vaira S, Yang C, McCoy A, Keys K, Xue S, Weinstein EJ, Novack DV, Cui X. Creation and preliminary characterization of a leptin knockout rat. Endocrinology. 2012;153:5622-5628.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 31]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
75.  Gafni O, Weinberger L, Mansour AA, Manor YS, Chomsky E, Ben-Yosef D, Kalma Y, Viukov S, Maza I, Zviran A. Derivation of novel human ground state naive pluripotent stem cells. Nature. 2013;504:282-286.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 764]  [Cited by in F6Publishing: 774]  [Article Influence: 70.4]  [Reference Citation Analysis (0)]