Network


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

Hotspot


Dive into the research topics where Taro Ishiguro is active.

Publication


Featured researches published by Taro Ishiguro.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Spinocerebellar ataxia type 6 knockin mice develop a progressive neuronal dysfunction with age-dependent accumulation of mutant CaV2.1 channels

Kei Watase; Curtis F. Barrett; Taisuke Miyazaki; Taro Ishiguro; Kinya Ishikawa; Yuanxin Hu; Toshinori Unno; Yaling Sun; Sayumi Kasai; Masahiko Watanabe; Christopher M. Gomez; Hidehiro Mizusawa; Richard W. Tsien; Huda Y. Zoghbi

Spinocerebellar ataxia type 6 (SCA6) is a neurodegenerative disorder caused by CAG repeat expansions within the voltage-gated calcium (CaV) 2.1 channel gene. It remains controversial whether the mutation exerts neurotoxicity by changing the function of CaV2.1 channel or through a gain-of-function mechanism associated with accumulation of the expanded polyglutamine protein. We generated three strains of knockin (KI) mice carrying normal, expanded, or hyperexpanded CAG repeat tracts in the Cacna1a locus. The mice expressing hyperexpanded polyglutamine (Sca684Q) developed progressive motor impairment and aggregation of mutant CaV2.1 channels. Electrophysiological analysis of cerebellar Purkinje cells revealed similar Ca2+ channel current density among the three KI models. Neither voltage sensitivity of activation nor inactivation was altered in the Sca684Q neurons, suggesting that expanded CAG repeat per se does not affect the intrinsic electrophysiological properties of the channels. The pathogenesis of SCA6 is apparently linked to an age-dependent process accompanied by accumulation of mutant CaV2.1 channels.


Neuron | 2017

Regulatory Role of RNA Chaperone TDP-43 for RNA Misfolding and Repeat-Associated Translation in SCA31

Taro Ishiguro; Nozomu Sato; Morio Ueyama; Nobuhiro Fujikake; Chantal Sellier; Akemi Kanegami; Eiichi Tokuda; Bita Zamiri; Terence Gall-Duncan; Mila Mirceta; Yoshiaki Furukawa; Takanori Yokota; Keiji Wada; J. Paul Taylor; Christopher E. Pearson; Nicolas Charlet-Berguerand; Hidehiro Mizusawa; Yoshitaka Nagai; Kinya Ishikawa

Microsatellite expansion disorders are pathologically characterized by RNA foci formation and repeat-associated non-AUG (RAN) translation. However, their underlying pathomechanisms and regulation of RAN translation remain unknown. We report that expression of expanded UGGAA (UGGAAexp) repeats, responsible for spinocerebellar ataxia type 31 (SCA31) in Drosophila, causes neurodegeneration accompanied by accumulation of UGGAAexp RNA foci and translation of repeat-associated pentapeptide repeat (PPR) proteins, consistent with observations in SCA31 patient brains. We revealed that motor-neuron disease (MND)-linked RNA-binding proteins (RBPs), TDP-43, FUS, and hnRNPA2B1, bind to and induce structural alteration of UGGAAexp. These RBPs suppress UGGAAexp-mediated toxicity in Drosophila by functioning as RNA chaperones for proper UGGAAexp folding and regulation of PPR translation. Furthermore, nontoxic short UGGAA repeat RNA suppressed mutated RBP aggregation and toxicity in MND Drosophila models. Thus, functional crosstalk of the RNA/RBP network regulates their own quality and balance, suggesting convergence of pathomechanisms in microsatellite expansion disorders and RBP proteinopathies.


Journal of Human Genetics | 2007

Redefining the disease locus of 16q22.1-linked autosomal dominant cerebellar ataxia

Takeshi Amino; Kinya Ishikawa; Shuta Toru; Taro Ishiguro; Nozomu Sato; Taiji Tsunemi; Miho Murata; Kazuhiro Kobayashi; Johji Inazawa; Tatsushi Toda; Hidehiro Mizusawa

AbstractThe 16q22.1-linked autosomal dominant cerebellar ataxia (16q-ADCA; Online Mendelian Inheritance in Man [OMIN] #117210) is one of the most common ADCAs in Japan. Previously, we had reported that the patients share a common haplotype by founder effect and that a C-to-T substitution (−16C>T) in the puratrophin-1 gene was strongly associated with the disease. However, recently, an exceptional patient without the substitution was reported, indicating that a true pathogenic mutation might be present elsewhere. In this study, we clarified the disease locus more definitely by the haplotype analysis of families showing pure cerebellar ataxia. In addition to microsatellite markers, the single nucleotide polymorphisms (SNPs) that we identified on the disease chromosome were examined to confirm the borders of the disease locus. The analysis of 64 families with the −16C>T substitution in the puratrophin-1 gene revealed one family showing an ancestral recombination event between SNP04 and SNP05 on the disease chromosome. The analysis of 22 families without identifiable genetic mutations revealed another family carrying the common haplotype centromeric to the puratrophin-1 gene, but lacking the −16C>T substitution in this gene. We concluded that the disease locus of 16q-ADCA was definitely confined to a 900-kb genomic region between the SNP04 and the −16C>T substitution in the puratrophin-1 gene in 16q22.1.


PLOS ONE | 2013

Cytoplasmic location of α1A voltage-gated calcium channel C-terminal fragment (Cav2.1-CTF) aggregate is sufficient to cause cell death.

Makoto Takahashi; Masato Obayashi; Taro Ishiguro; Nozomu Sato; Yusuke Niimi; Kokoro Ozaki; Kaoru Mogushi; Yasen Mahmut; Hiroshi Tanaka; Fuminori Tsuruta; Ricardo E. Dolmetsch; Mitsunori Yamada; Hitoshi Takahashi; Takeo Kato; Osamu Mori; Yoshinobu Eishi; Hidehiro Mizusawa; Kinya Ishikawa

The human α1A voltage-dependent calcium channel (Cav2.1) is a pore-forming essential subunit embedded in the plasma membrane. Its cytoplasmic carboxyl(C)-tail contains a small poly-glutamine (Q) tract, whose length is normally 4∼19 Q, but when expanded up to 20∼33Q, the tract causes an autosomal-dominant neurodegenerative disorder, spinocerebellar ataxia type 6 (SCA6). A recent study has shown that a 75-kDa C-terminal fragment (CTF) containing the polyQ tract remains soluble in normal brains, but becomes insoluble mainly in the cytoplasm with additional localization to the nuclei of human SCA6 Purkinje cells. However, the mechanism by which the CTF aggregation leads to neurodegeneration is completely elusive, particularly whether the CTF exerts more toxicity in the nucleus or in the cytoplasm. We tagged recombinant (r)CTF with either nuclear-localization or nuclear-export signal, created doxycyclin-inducible rat pheochromocytoma (PC12) cell lines, and found that the CTF is more toxic in the cytoplasm than in the nucleus, the observations being more obvious with Q28 (disease range) than with Q13 (normal-length). Surprisingly, the CTF aggregates co-localized both with cAMP response element-binding protein (CREB) and phosphorylated-CREB (p-CREB) in the cytoplasm, and Western blot analysis showed that the quantity of CREB and p-CREB were both decreased in the nucleus when the rCTF formed aggregates in the cytoplasm. In human brains, polyQ aggregates also co-localized with CREB in the cytoplasm of SCA6 Purkinje cells, but not in other conditions. Collectively, the cytoplasmic Cav2.1-CTF aggregates are sufficient to cause cell death, and one of the pathogenic mechanisms may be abnormal CREB trafficking in the cytoplasm and reduced CREB and p-CREB levels in the nuclei.


Neuropathology | 2012

Reduced brain-derived neurotrophic factor (BDNF) mRNA expression and presence of BDNF-immunoreactive granules in the spinocerebellar ataxia type 6 (SCA6) cerebellum

Makoto Takahashi; Kinya Ishikawa; Nozomu Sato; Masato Obayashi; Yusuke Niimi; Taro Ishiguro; Mitsunori Yamada; Yasuko Toyoshima; Hitoshi Takahashi; Takeo Kato; Masaki Takao; Shigeo Murayama; Osamu Mori; Yoshinobu Eishi; Hidehiro Mizusawa

Spinocerebellar ataxia type 6 (SCA6) is an autosomal‐dominant neurodegenerative disorder caused by a small expansion of tri‐nucleotide (CAG) repeat encoding polyglutamine (polyQ) in the gene for α1A voltage‐dependent calcium channel (Cav2.1). Thus, this disease is one of the nine neurodegenerative disorders called polyQ diseases. The Purkinje cell predominant neuronal loss is the characteristic neuropathology of SCA6, and a 75‐kDa carboxy‐terminal fragment (CTF) of Cav2.1 containing polyQ, which remains soluble in normal brains, becomes insoluble in the cytoplasm of SCA6 Purkinje cells. Because the suppression of the brain‐derived neurotrophic factor (BDNF) expression is a potentially momentous phenomenon in many other polyQ diseases, we implemented BDNF expression analysis in SCA6 human cerebellum using quantitative RT‐PCR for the BDNF mRNA, and by immunohistochemistry for the BDNF protein. We observed significantly reduced BDNF mRNA levels in SCA6 cerebellum (n = 3) compared to controls (n = 6) (Mann–Whitney U‐test, P = 0.0201). On immunohistochemistry, BDNF protein was only weakly stained in control cerebellum. On the other hand, we found numerous BDNF‐immunoreactive granules in dendrites of SCA6 Purkinje cells. We did not observe similar BDNF‐immunoreactive granules in other polyQ diseases, such as Huntingtons disease or SCA2. As we often observed that the 1C2‐positive Cav2.1 aggregates existed more proximally than the BDNF‐positive granules in the dendrites, we speculated that the BDNF protein trafficking in dendrites may be disturbed by Cav2.1 aggregates in SCA6 Purkinje cells. We conclude that the SCA6 pathogenic mechanism associates with the BDNF mRNA expression reduction and abnormal localization of BDNF protein.


Clinical Neurology and Neurosurgery | 2013

Paradoxical cerebral embolism with patent foramen ovale and deep venous thrombosis caused by a massive myoma uteri

Shuta Toru; Toyonari Murata; Maya Ohara; Taro Ishiguro; Takayoshi Kobayashi

A 42-year-old dextral woman came to our hospital with right emiparesis. A huge uteral myoma was found 10 years ago, and he regularly visited our gynecology department. She had had neiher other past illnesses nor the habit of drinking or smoking. Drug istory for oral contraceptives is negative. At age 41, she suddenly uffered left hemiparesis and was admitted to another hospital. erebral infarction was diagnosed in the right middle cerebral rtery (MCA) area. Brain angiography showed stenosis of the M2 ortion of the right MCA, but no atherosclerotic changes. She was ischarged with no neurological symptoms, after which cilostazol ad been prescribed. On admission to our hospital, right hemiparesis and severe otor aphasia were present, and there was slight edema in her left eg. Neurological and general findings showed no other abnormaliies. d-Dimer was elevated to 30.8 g/ml (normal range < 1 g/ml), ut other laboratory data on admission for cholesterol, HbA1c,


Journal of Human Genetics | 2008

Direct and accurate measurement of CAG repeat configuration in the ataxin-1 (ATXN-1) gene by “dual-fluorescence labeled PCR-restriction fragment length analysis”

Jiang X. Lin; Kinya Ishikawa; Masaki Sakamoto; Taiji Tsunemi; Taro Ishiguro; Takeshi Amino; Shuta Toru; Ikuko Kondo; Hidehiro Mizusawa

AbstractSpinocerebellar ataxia type 1 (SCA1; OMIM: #164400) is an autosomal dominant cerebellar ataxia caused by an expansion of CAG repeat, which encodes polyglutamine, in the ataxin-1 (ATXN1) gene. Length of polyglutamine in the ATXN1 protein is the critical determinant of pathogenesis of this disease. Molecular diagnosis of SCA1 is usually undertaken by assessing the length of CAG repeat configuration using primers spanning this configuration. However, this conventional method may potentially lead to misdiagnosis in assessing polyglutamine-encoding CAG repeat length, since CAT interruptions may be present within the CAG repeat configuration, not only in normal controls but also in neurologically symptomatic subjects. We developed a new method for assessing actual CAG repeat numbers not interrupted by CAT sequences. Polymerase chain reaction using a primer pair labeled with two different fluorescences followed by restriction enzyme digestion with SfaNI which recognizes the sequence “GCATC(N)5”, lengths of actual CAG repeats that encode polyglutamine were directly detected. We named this method “dual fluorescence labeled PCR-restriction fragment length analysis”. We found that numbers of actual CAG repeat encoding polyglutamine do not overlap between our cohorts of normal chromosomes (n = 385) and SCA1 chromosomes (n = 5). We conclude that the present method is a useful way for molecular diagnosis of SCA1.


American Journal of Human Genetics | 2009

Spinocerebellar ataxia type 31 is associated with "inserted" penta-nucleotide repeats containing (TGGAA)n.

Nozomu Sato; Takeshi Amino; Kazuhiro Kobayashi; Shuichi Asakawa; Taro Ishiguro; Taiji Tsunemi; Makoto Takahashi; Tohru Matsuura; Kevin M. Flanigan; Sawa Iwasaki; Fumitoshi Ishino; Yuko Saito; Shigeo Murayama; Mari Yoshida; Yoshio Hashizume; Yuji Takahashi; Shoji Tsuji; Nobuyoshi Shimizu; Tatsushi Toda; Kinya Ishikawa; Hidehiro Mizusawa


Acta Neuropathologica | 2010

The carboxy-terminal fragment of α1A calcium channel preferentially aggregates in the cytoplasm of human spinocerebellar ataxia type 6 Purkinje cells

Taro Ishiguro; Kinya Ishikawa; Makoto Takahashi; Masato Obayashi; Takeshi Amino; Nozomu Sato; Masaki Sakamoto; Hiroto Fujigasaki; Fuminori Tsuruta; Ricardo E. Dolmetsch; Takao Arai; Hidenao Sasaki; Kazuro Nagashima; Takeo Kato; Mitsunori Yamada; Hitoshi Takahashi; Yoshio Hashizume; Hidehiro Mizusawa


Neuropathology | 2013

Abnormal RNA structures (RNA foci) containing a penta‐nucleotide repeat (UGGAA)n in the Purkinje cell nucleus is associated with spinocerebellar ataxia type 31 pathogenesis

Yusuke Niimi; Makoto Takahashi; Emiko Sugawara; Shigeaki Umeda; Masato Obayashi; Nozomu Sato; Taro Ishiguro; Miwa Higashi; Yoshinobu Eishi; Hidehiro Mizusawa; Kinya Ishikawa

Collaboration


Dive into the Taro Ishiguro's collaboration.

Top Co-Authors

Avatar

Hidehiro Mizusawa

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Kinya Ishikawa

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Nozomu Sato

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Makoto Takahashi

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Takeshi Amino

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Masato Obayashi

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Shuta Toru

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Taiji Tsunemi

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge