Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Tomoji Mashimo is active.

Publication


Featured researches published by Tomoji Mashimo.


Nature Genetics | 2008

Progress and prospects in rat genetics: a community view

Timothy J. Aitman; John K. Critser; Edwin Cuppen; Anna F. Dominiczak; Xosé M. Fernández-Suárez; Jonathan Flint; Dominique Gauguier; Aron M. Geurts; Michael N. Gould; Peter C. Harris; Rikard Holmdahl; Norbert Hubner; Zsuzsanna Izsvák; Howard J. Jacob; Takashi Kuramoto; Anne E. Kwitek; Anna Marrone; Tomoji Mashimo; Carol Moreno; John J. Mullins; Linda J. Mullins; Tomas Olsson; Michal Pravenec; Lela K. Riley; Kathrin Saar; Tadao Serikawa; James D Shull; Claude Szpirer; Simon N. Twigger; Birger Voigt

The rat is an important system for modeling human disease. Four years ago, the rich 150-year history of rat research was transformed by the sequencing of the rat genome, ushering in an era of exceptional opportunity for identifying genes and pathways underlying disease phenotypes. Genome-wide association studies in human populations have recently provided a direct approach for finding robust genetic associations in common diseases, but identifying the precise genes and their mechanisms of action remains problematic. In the context of significant progress in rat genomic resources over the past decade, we outline achievements in rat gene discovery to date, show how these findings have been translated to human disease, and document an increasing pace of discovery of new disease genes, pathways and mechanisms. Finally, we present a set of principles that justify continuing and strengthening genetic studies in the rat model, and further development of genomic infrastructure for rat research.


PLOS ONE | 2010

Generation of Knockout Rats with X-Linked Severe Combined Immunodeficiency (X-SCID) Using Zinc-Finger Nucleases

Tomoji Mashimo; Akiko Takizawa; Birger Voigt; Kazuto Yoshimi; Hiroshi Hiai; Takashi Kuramoto; Tadao Serikawa

Background Although the rat is extensively used as a laboratory model, the inability to utilize germ line-competent rat embryonic stem (ES) cells has been a major drawback for studies that aim to elucidate gene functions. Recently, zinc-finger nucleases (ZFNs) were successfully used to create genome-specific double-stranded breaks and thereby induce targeted gene mutations in a wide variety of organisms including plants, drosophila, zebrafish, etc. Methodology/Principal Findings We report here on ZFN-induced gene targeting of the rat interleukin 2 receptor gamma (Il2rg) locus, where orthologous human and mouse mutations cause X-linked severe combined immune deficiency (X-SCID). Co-injection of mRNAs encoding custom-designed ZFNs into the pronucleus of fertilized oocytes yielded genetically modified offspring at rates greater than 20%, which possessed a wide variety of deletion/insertion mutations. ZFN-modified founders faithfully transmitted their genetic changes to the next generation along with the severe combined immune deficiency phenotype. Conclusions and Significance The efficient and rapid generation of gene knockout rats shows that using ZFN technology is a new strategy for creating gene-targeted rat models of human diseases. In addition, the X-SCID rats that were established in this study will be valuable in vivo tools for evaluating drug treatment or gene therapy as well as model systems for examining the treatment of xenotransplanted malignancies.


Nature Genetics | 2008

An ENU-induced mutant archive for gene targeting in rats

Tomoji Mashimo; Katsuhiko Yanagihara; Satoko Tokuda; Birger Voigt; Akiko Takizawa; Reiko Nakajima; Megumi Kato; Masumi Hirabayashi; Takashi Kuramoto; Tadao Serikawa

To the editor: Although the laboratory rat is increasingly being used as a mammalian model in biomedical research, no technology exists thus far for the production of in vivo genetically engineered mutations equivalent to knockout or knock-in mice because of the lack of development of functional embryonic stem cells in this species. Rat spermatogonial stem cells may possibly have greater potential use for genetic engineering to produce gene-targeted rats1,2. The use of somatic cell nuclear transfer to develop cloned rats as an alternative to using embryonic stem cells has also been reported3, but it seems to be a very difficult technique to perform. At present, N-ethyl-Nnitrosourea (ENU) mutagenesis followed by a screening method to detect single-nucleotide substitutions within the targeted gene seems to be the most promising technology in rats, as previously reported by several groups4,5,6. The screening protocol, however, either a yeastbased screening assay4,5 or a highthroughput resequencing-based screening technique7, is expensive. In addition, gene-driven ENU mutagenesis approaches in rats are not a very efficient use of resources because most of the rats generated are usually discarded within a few weeks or months after the targeted genes have been screened owing to a lack of reliable sperm conservation and rederivation technologies. We have developed a new, efficient approach that combines two methods: a high-throughput, lowa b


Scientific Reports | 2013

Efficient gene targeting by TAL effector nucleases coinjected with exonucleases in zygotes

Tomoji Mashimo; Takehito Kaneko; Tetsushi Sakuma; Junya Kobayashi; Yayoi Kunihiro; Birger Voigt; Takashi Yamamoto; Tadao Serikawa

TAL Effector Nucleases (TALENs) are versatile tools for targeted gene editing in various species. However, their efficiency is still insufficient, especially in mammalian embryos. Here, we showed that combined expression of Exonuclease 1 (Exo1) with engineered site-specific TALENs provided highly efficient disruption of the endogenous gene in rat fibroblast cells. A similar increased efficiency of up to ~30% with Exo1 was also observed in fertilized rat eggs, and in the production of knockout rats for the albino (Tyr) gene. These findings demonstrate TALENs with Exo1 is an easy and efficient method of generating gene knockouts using zygotes, which increases the range of gene targeting technologies available to various species.


The Journal of Neuroscience | 2010

A Missense Mutation of the Gene Encoding Voltage-Dependent Sodium Channel (Nav1.1) Confers Susceptibility to Febrile Seizures in Rats

Tomoji Mashimo; Iori Ohmori; Mamoru Ouchida; Yukihiro Ohno; Toshiko Tsurumi; Takafumi Miki; Minoru Wakamori; Shizuka Ishihara; Takashi Yoshida; Akiko Takizawa; Megumi Kato; Masumi Hirabayashi; Masashi Sasa; Yasuo Mori; Tadao Serikawa

Although febrile seizures (FSs) are the most common convulsive syndrome in infants and childhood, the etiology of FSs has remained unclarified. Several missense mutations of the Nav1.1 channel (SCN1A), which alter channel properties, have been reported in a familial syndrome of GEFS+ (generalized epilepsy with febrile seizures plus). Here, we generated Scn1a-targeted rats carrying a missense mutation (N1417H) in the third pore region of the sodium channel by gene-driven ENU (N-ethyl-N-nitrosourea) mutagenesis. Despite their normal appearance under ordinary circumstances, Scn1a mutant rats exhibited remarkably high susceptibility to hyperthermia-induced seizures, which involve generalized clonic and/or tonic–clonic convulsions with paroxysmal epileptiform discharges. Whole-cell patch-clamp recordings from HEK cells expressing N1417H mutant channels and from hippocampal GABAergic interneurons of N1417H mutant rats revealed a significant shift of the inactivation curve in the hyperpolarizing direction. In addition, clamp recordings clearly showed the reduction in action potential amplitude in the hippocampal interneurons of these rats. These findings suggest that a missense mutation (N1417H) of the Nav1.1 channel confers susceptibility to FS and the impaired biophysical properties of inhibitory GABAergic neurons underlie one of the mechanisms of FS.


Cell Reports | 2012

Generation and Characterization of Severe Combined Immunodeficiency Rats

Tomoji Mashimo; Akiko Takizawa; Junya Kobayashi; Yayoi Kunihiro; Kazuto Yoshimi; Saeko Ishida; Koji Tanabe; Ami Yanagi; Asato Tachibana; Jun Hirose; Jun-ichiro Yomoda; Shiho Morimoto; Takashi Kuramoto; Birger Voigt; Takeshi Watanabe; Hiroshi Hiai; Chise Tateno; Kenshi Komatsu; Tadao Serikawa

Severe combined immunodeficiency (SCID) mice, the most widely used animal model of DNA-PKcs (Prkdc) deficiency, have contributed enormously to our understanding of immunodeficiency, lymphocyte development, and DNA-repair mechanisms, and they are ideal hosts for allogeneic and xenogeneic tissue transplantation. Here, we use zinc-finger nucleases to generate rats that lack either the Prkdc gene (SCID) or the Prkdc and Il2rg genes (referred to as F344-scid gamma [FSG] rats). SCID rats show several phenotypic differences from SCID mice, including growth retardation, premature senescence, and a more severe immunodeficiency without leaky phenotypes. Double-knockout FSG rats show an even more immunocompromised phenotype, such as the abolishment of natural killer cells. Finally, xenotransplantation of human induced pluripotent stem cells, ovarian cancer cells, and hepatocytes shows that SCID and FSG rats can act as hosts for xenogeneic tissue grafts and stem cell transplantation and may be useful for preclinical testing of new drugs.


Cancer Science | 2009

Enhanced colitis-associated colon carcinogenesis in a novel Apc mutant rat

Kazuto Yoshimi; Takuji Tanaka; Akiko Takizawa; Megumi Kato; Masumi Hirabayashi; Tomoji Mashimo; Tadao Serikawa; Takashi Kuramoto

To establish an efficient rat model for colitis‐associated colorectal cancer, azoxymethane and dextran sodium sulfate (AOM/DSS)‐induced colon carcinogenesis was applied to a novel adenomatous polyposis coli (Apc) mutant, the Kyoto Apc Delta (KAD) rat. The KAD rat was derived from ethylnitrosourea mutagenesis and harbors a nonsense mutation in the Apc gene (S2523X). The truncated APC of the KAD rat was deduced to lack part of the basic domain, an EB1‐binding domain, and a PDZ domain, but retained an intact β‐catenin binding region. KAD rats, homozygous for the Apc mutation on a genetic background of the F344 rat, showed no spontaneous tumors in the gastrointestinal tract. At 5u2003weeks of age, male KAD rats were given a single subcutaneous administration of AOM (20u2003mg/kg, bodyweight). One week later, they were given DSS (2% in drinking water) for 1u2003week. At week 15, the incidence and multiplicity of colon tumors developed in the KAD rat were remarkably severe compared with those in the F344 rat: 100 versus 50% in incidence and 10.7u2003±u20033.5 versus 0.8u2003±u20031.0 in multiplicity. KAD tumors were dominantly distributed in the rectum and distal colon, resembling human colorectal cancer. Accumulation of β‐catenin protein and frequent β‐catenin mutations were prominent features of KAD colon tumors. To our knowledge, AOM/DSS‐induced colon carcinogenesis using the KAD rat is the most efficient to induce colon tumors in the rat, and therefore would be available as an excellent model for human colitis‐associated CRC.


Neurobiology of Disease | 2011

Scn1a missense mutation causes limbic hyperexcitability and vulnerability to experimental febrile seizures.

Yukihiro Ohno; Shizuka Ishihara; Tomoji Mashimo; Nobumasa Sofue; Saki Shimizu; Takuji Imaoku; Toshiko Tsurumi; Masashi Sasa; Tadao Serikawa

Mutations of the voltage-gated sodium (Na(v)) channel subunit SCN1A have been implicated in the pathogenesis of human febrile seizures including generalized epilepsy with febrile seizures plus (GEFS+) and severe myoclonic epilepsy in infancy (SMEI). Hyperthermia-induced seizure-susceptible (Hiss) rats are the novel rat model carrying a missense mutation (N1417H) of Scn1a, which is located in the third pore-forming region of the Na(v)1.1 channel. Here, we conducted behavioral and neurochemical studies to clarify the functional relevance of the Scn1a mutation in vivo and the mechanism underlying the vulnerability to hyperthermic seizures. Hiss rats showed markedly high susceptibility to hyperthermic seizures (mainly generalized clonic seizures) which were synchronously associated with paroxysmal epileptiform discharges. Immunohistochemical analysis of brain Fos expression revealed that hyperthermic seizures induced a widespread elevation of Fos-immunoreactivity in the cerebral cortices including the motor area, piriform, and insular cortex. In the subcortical regions, hyperthermic seizures enhanced Fos expression region--specifically in the limbic and paralimbic regions (e.g., hippocampus, amygdala, and perirhinal-entorhinal cortex) without affecting other brain regions (e.g., basal ganglia, diencephalon, and lower brainstem), suggesting a primary involvement of limbic system in the induction of hyperthermic seizures. In addition, Hiss rats showed a significantly lower threshold than the control animals in inducing epileptiform discharges in response to local stimulation of the hippocampus (hippocampal afterdischarges). Furthermore, hyperthermic seizures in Hiss rats were significantly alleviated by the antiepileptic drugs, diazepam and sodium valproate, while phenytoin or ethosuximide were ineffective. The present findings support the notion that Hiss rats are useful as a novel rat model of febrile seizures and suggest that hyperexcitability of limbic neurons associated with Scn1a missense mutation plays a crucial role in the pathogenesis of febrile seizures.


Human Molecular Genetics | 2012

A rat model for LGI1-related epilepsies

Stéphanie Baulac; Saeko Ishida; Tomoji Mashimo; Morgane Boillot; Naohiro Fumoto; Mitsuru Kuwamura; Yukihiro Ohno; Akiko Takizawa; Toshihiro Aoto; Masatsugu Ueda; Akio Ikeda; Eric LeGuern; Ryosuke Takahashi; Tadao Serikawa

Mutations of the leucine-rich glioma-inactivated 1 (LGI1) gene cause an autosomal dominant partial epilepsy with auditory features also known as autosomal-dominant lateral temporal lobe epilepsy. LGI1 is also the main antigen present in sera and cerebrospinal fluids of patients with limbic encephalitis and seizures, highlighting its importance in a spectrum of epileptic disorders. LGI1 encodes a neuronal secreted protein, whose brain function is still poorly understood. Here, we generated, by ENU (N-ethyl-N-nitrosourea) mutagenesis, Lgi1-mutant rats carrying a missense mutation (L385R). We found that the L385R mutation prevents the secretion of Lgi1 protein by COS7 transfected cells. However, the L385R-Lgi1 protein was found at low levels in the brains and cultured neurons of Lgi1-mutant rats, suggesting that mutant protein may be destabilized in vivo. Studies on the behavioral phenotype and intracranial electroencephalographic signals from Lgi1-mutant rats recalled several features of the human genetic disorder. We show that homozygous Lgi1-mutant rats (Lgi1(L385R/L385R)) generated early-onset spontaneous epileptic seizures from P10 and died prematurely. Heterozygous Lgi1-mutant rats (Lgi1(+/L385R)) were more susceptible to sound-induced, generalized tonic-clonic seizures than control rats. Audiogenic seizures were suppressed by antiepileptic drugs such as carbamazepine, phenytoin and levetiracetam, which are commonly used to treat partial seizures, but not by the prototypic absence seizure drug, ethosuximide. Our findings provide the first rat model with a missense mutation in Lgi1 gene, an original model complementary to knockout mice. This study revealed that LGI1 disease-causing missense mutations might cause a depletion of the protein in neurons, and not only a failure of Lgi1 secretion.


Current Pharmaceutical Biotechnology | 2009

Rat resources in biomedical research.

Tomoji Mashimo; Tadao Serikawa

The laboratory rat is obviously an important model for physiology, pathology, pharmacology, toxicology, and transplantation experiments. The value for pharmacological research is immense since virtually every drug approved for human treatment passes through the body of laboratory rats. Hundreds of unique rat models have been developed to mimic pathological and physiological human clinical conditions, especially in the case of complex diseases. Many of the model rats are deposited into rat resource centers, from which researchers can use and share animals and rat related resources in biomedical research. Recent progressing technologies for genetically engineered rats, such as traditional transgenesis, chemical ENU mutagenesis, and transposon insertional mutagenesis, will provide thousands of useful rat models for functional genomics and human diseases. Globally acting rat resource centers are prerequisites for successful and sustainable research in the biomedical field where the rats are used as model species.

Collaboration


Dive into the Tomoji Mashimo's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yukihiro Ohno

Osaka University of Pharmaceutical Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Masumi Hirabayashi

Graduate University for Advanced Studies

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mitsuru Kuwamura

Osaka Prefecture University

View shared research outputs
Researchain Logo
Decentralizing Knowledge