Updated on 2024/05/15

写真a

 
Ishii Hirotaka
 
Affiliation
Graduate School of Medicine, Department of Anatomy and Neurobiology, Graduate Shool Professor
Faculty of Medicine, Department of Anatomy and Neurobiology, Graduate Shool Professor
Title
Graduate Shool Professor
External link

Degree

  • Ph.D. ( 2006.3   The University of Tokyo )

Research Interests

  • Estrogen

  • Chemotherapy resistance

  • 形態機能学

  • Reproductive endocrinology

  • Neuroendocrinology

Research Areas

  • Life Science / Anatomy

Education

  • The University of Tokyo   Graduate School of Science   Physics

    2003.4 - 2006.3

      More details

  • The University of Tokyo   Graduate School of Science   Physics

    2001.4 - 2003.3

      More details

  • The University of Tokyo   Department of Science   Physics

    1999.4 - 2001.3

      More details

  • The University of Tokyo   Department of Art and Sciences   Science Cource I

    1997.4 - 1999.3

      More details

Research History

  • Nippon Medical School Graduate School of Medicine   Department of Anatomy and Neurobiology   Graduate School Professor

    2022.4

      More details

    Country:Japan

    researchmap

  • Nippon Medical School Graduate School of Medicine   Department of Anatomy and Neurobiology   Associate Professor

    2018.4 - 2022.3

      More details

    Country:Japan

    researchmap

  • Nippon Medical School, Graduate School of Medicine   Department of Anatomy and Neurobiology   Senior Assistant Professor

    2013.4 - 2018.3

      More details

  • Nippon Medical School, Graduate School of Medicine   Department of Anatomy and Neurobiology   Assistant Professor

    2012.4 - 2013.3

      More details

  • Nippon Medical School, Graduate School of Medicine   Department of Physiology   Assistant Professor

    2010.4 - 2012.3

      More details

  • Nippon Medical School, Graduate School of Medicine   Department of Physiology   Postdoctor

    2006.4 - 2010.3

      More details

▼display all

Professional Memberships

▼display all

Committee Memberships

  • 日本組織細胞化学会   評議員  

    2023.1   

      More details

  • 日本解剖学会   代議員  

    2022.8   

      More details

  • 日本神経内分泌学会   評議員  

    2012.4   

      More details

  • 日本生理学会   評議員  

    2012.3   

      More details

Papers

  • Transcriptomic Profiling of Lactotroph Pituitary Neuroendocrine Tumors via RNA Sequencing and Ingenuity Pathway Analysis. International journal

    Yujiro Hattori, Shigeyuki Tahara, Hitoshi Ozawa, Akio Morita, Hirotaka Ishii

    Neuroendocrinology   2024.4

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    INTRODUCTION: Lactotroph pituitary neuroendocrine tumors (PitNETs) are common pituitary tumors, but their underlying molecular mechanisms remain unclear. This study aimed to investigate the transcriptomic landscape of lactotroph PitNETs and identify potential molecular mechanisms and therapeutic targets through RNA sequencing and ingenuity pathway analysis (IPA). METHODS: Lactotroph PitNET tissues from five surgical cases without dopamine agonist treatment underwent RNA sequencing. Normal pituitary tissues from three patients served as controls. Differentially expressed genes (DEGs) were identified, and the functional pathways and gene networks were explored by IPA. RESULTS: Transcriptome analysis revealed that lactotroph PitNETs had gene expression patterns that were distinct from normal pituitary tissues. We identified 1,172 upregulated DEGs, including nine long intergenic noncoding RNAs (lincRNAs) belonging to the top 30 DEGs. IPA of the upregulated DEGs showed that the estrogen receptor signaling, oxidative phosphorylation signaling, and EIF signaling were activated. In gene network analysis, key upstream regulators, such as EGR1, PRKACA, PITX2, CREB1, and JUND, may play critical roles in lactotroph PitNETs. CONCLUSION: This study provides a comprehensive transcriptomic profile of lactotroph PitNETs and highlights the potential involvement of lincRNAs and specific signaling pathways in tumor pathogenesis. The identified upstream regulators may be potential therapeutic targets for future investigations.

    DOI: 10.1159/000539017

    PubMed

    researchmap

  • Endothelial cells regulate alveolar morphogenesis by constructing basement membranes acting as a scaffold for myofibroblasts. Reviewed International journal

    Haruko Watanabe-Takano, Katsuhiro Kato, Eri Oguri-Nakamura, Tomohiro Ishii, Koji Kobayashi, Takahisa Murata, Koichiro Tsujikawa, Takaki Miyata, Yoshiaki Kubota, Yasuyuki Hanada, Koichi Nishiyama, Tetsuro Watabe, Reinhard Fässler, Hirotaka Ishii, Naoki Mochizuki, Shigetomo Fukuhara

    Nature communications   15 ( 1 )   1622 - 1622   2024.3

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Alveologenesis is a spatially coordinated morphogenetic event, during which alveolar myofibroblasts surround the terminal sacs constructed by epithelial cells and endothelial cells (ECs), then contract to form secondary septa to generate alveoli in the lungs. Recent studies have demonstrated the important role of alveolar ECs in this morphogenetic event. However, the mechanisms underlying EC-mediated alveologenesis remain unknown. Herein, we show that ECs regulate alveologenesis by constructing basement membranes (BMs) acting as a scaffold for myofibroblasts to induce septa formation through activating mechanical signaling. Rap1, a small GTPase of the Ras superfamily, is known to stimulate integrin-mediated cell adhesions. EC-specific Rap1-deficient (Rap1iECKO) mice exhibit impaired septa formation and hypo-alveolarization due to the decreased mechanical signaling in myofibroblasts. In Rap1iECKO mice, ECs fail to stimulate integrin β1 to recruit Collagen type IV (Col-4) into BMs required for myofibroblast-mediated septa formation. Consistently, EC-specific integrin β1-deficient mice show hypo-alveolarization, defective mechanical signaling in myofibroblasts, and disorganized BMs. These data demonstrate that alveolar ECs promote integrin β1-mediated Col-4 recruitment in a Rap1-dependent manner, thereby constructing BMs acting as a scaffold for myofibroblasts to induce mechanical signal-mediated alveologenesis. Thus, this study unveils a mechanism of organ morphogenesis mediated by ECs through intrinsic functions.

    DOI: 10.1038/s41467-024-45910-y

    PubMed

    researchmap

  • Chronic estradiol exposure suppresses luteinizing hormone surge without affecting kisspeptin neurons and estrogen receptor alpha in anteroventral periventricular nucleus. Reviewed International journal

    Yuyu Kunimura, Kinuyo Iwata, Hirotaka Ishii, Hitoshi Ozawa

    Biology of reproduction   110 ( 1 )   90 - 101   2024.1

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Mammalian ovulation is induced by a luteinizing hormone surge, which is triggered by elevated plasma estrogen levels; however, chronic exposure to high levels of estradiol is known to inhibit luteinizing hormone secretion. In the present study, we hypothesized that the inhibition of the luteinizing hormone surge by chronic estradiol exposure is due to the downregulation of the estrogen receptor alpha in kisspeptin neurons at hypothalamic anteroventral periventricular nucleus, which is known as the gonadotropin-releasing hormone/luteinizing hormone surge generator. Animals exposed to estradiol for 2 days showed an luteinizing hormone surge, whereas those exposed for 14 days showed a significant suppression of luteinizing hormone. Chronic estradiol exposure did not affect the number of kisspeptin neurons and the percentage of kisspeptin neurons with estrogen receptor alpha or c-Fos in anteroventral periventricular nucleus, but it did affect the number of kisspeptin neurons in arcuate nucleus. Furthermore, chronic estradiol exposure did not affect gonadotropin-releasing hormone neurons. In the pituitary, 14-day estradiol exposure significantly reduced the expression of Lhb mRNA and LHβ-immunoreactive areas. Gonadotropin-releasing hormone-induced luteinizing hormone release was also reduced significantly by 14-day estradiol exposure. We revealed that the suppression of an luteinizing hormone surge by chronic estradiol exposure was induced in association with the significant reduction in kisspeptin neurons in arcuate nucleus, luteinizing hormone expression in the pituitary, and pituitary responsiveness to gonadotropin-releasing hormone, and this was not caused by changes in the estrogen receptor alpha-expressing kisspeptin neurons in anteroventral periventricular nucleus and gonadotropin-releasing hormone neurons, which are responsible for estradiol positive feedback.

    DOI: 10.1093/biolre/ioad129

    PubMed

    researchmap

  • Sex and interspecies differences in ESR2-expressing cell distributions in mouse and rat brains Reviewed

    Masahiro Morishita, Shimpei Higo, Kinuyo Iwata, Hirotaka Ishii

    Biology of Sex Differences   14 ( 1 )   2023.12

     More details

    Authorship:Last author, Corresponding author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:Springer Science and Business Media LLC  

    Abstract

    Background

    ESR2, a nuclear estrogen receptor also known as estrogen receptor β, is expressed in the brain and contributes to the actions of estrogen in various physiological phenomena. However, its expression profiles in the brain have long been debated because of difficulties in detecting ESR2-expressing cells. In the present study, we aimed to determine the distribution of ESR2 in rodent brains, as well as its sex and interspecies differences, using immunohistochemical detection with a well-validated anti-ESR2 antibody (PPZ0506).

    Methods

    To determine the expression profiles of ESR2 protein in rodent brains, whole brain sections from mice and rats of both sexes were subjected to immunostaining for ESR2. In addition, to evaluate the effects of circulating estrogen on ESR2 expression profiles, ovariectomized female mice and rats were treated with low or high doses of estrogen, and the resulting numbers of ESR2-immunopositive cells were analyzed. Welch’s t-test was used for comparisons between two groups for sex differences, and one-way analysis of variance followed by the Tukey–Kramer test were used for comparisons among multiple groups with different estrogen treatments.

    Results

    ESR2-immunopositive cells were observed in several subregions of mouse and rat brains, including the preoptic area, extended amygdala, hypothalamus, mesencephalon, and cerebral cortex. Their distribution profiles exhibited sex and interspecies differences. In addition, low-dose estrogen treatment in ovariectomized female mice and rats tended to increase the numbers of ESR2-immunopositive cells, whereas high-dose estrogen treatment tended to decrease these numbers.

    Conclusions

    Immunohistochemistry using the well-validated PPZ0506 antibody revealed a more localized expression of ESR2 protein in rodent brains than has previously been reported. Furthermore, there were marked sex and interspecies differences in its distribution. Our histological analyses also revealed estrogen-dependent changes in ESR2 expression levels in female brains. These findings will be helpful for understanding the ESR2-mediated actions of estrogen in the brain.

    DOI: 10.1186/s13293-023-00574-z

    researchmap

    Other Link: https://link.springer.com/article/10.1186/s13293-023-00574-z/fulltext.html

  • Impact of Well-validated Anti-human ERβ Monoclonal Antibody PPZ0506 on ERβ Research Reviewed

    Hirotaka Ishii, Yujiro Hattori, Shimpei Higo, Masahiro Morishita, Mina Ozawa, Mai Otsuka, Keisuke Matsumoto, Hitoshi Ozawa

    19 ( 4 )   332 - 338   2023.12

     More details

    Authorship:Lead author, Corresponding author   Language:Japanese   Publishing type:Research paper (scientific journal)  

    researchmap

  • Estrogen Receptor α Isoforms Generated by Alternative Use of Cryptic Exons Invited Reviewed

    Hirotaka Ishii, Yujiro Hattori, Hitoshi Ozawa

    Journal of Nippon Medical School   90 ( 5 )   364 - 371   2023.10

     More details

    Authorship:Lead author, Corresponding author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:Medical Association of Nippon Medical School  

    DOI: 10.1272/jnms.jnms.2023_90-507

    researchmap

  • Central injection of neuropeptide B induces luteinizing hormone release in male and female rats Reviewed

    Youki Watanabe, Kinuyo Iwata, Shiori Minabe, Nobuhiro Nakao, Hirotaka Ishii, Hitoshi Ozawa

    Peptides   171064 - 171064   2023.7

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:Elsevier BV  

    DOI: 10.1016/j.peptides.2023.171064

    researchmap

  • Recent Advances in High-sensitivity In Situ Hybridization and Costs and Benefits to Consider When Employing These Methods Invited Reviewed

    Shimpei Higo, Hirotaka Ishii, Hitoshi Ozawa

    ACTA HISTOCHEMICA ET CYTOCHEMICA   56 ( 3 )   49 - 54   2023.6

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:Japan Society of Histochemistry & Cytochemistry  

    DOI: 10.1267/ahc.23-00024

    researchmap

  • Immunohistochemistry for ESR2 with a Mouse Monoclonal Antibody (PPZ0506) Reviewed

    Masahiro Morishita, Shimpei Higo, Yujiro Hattori, Mina Ozawa, Mai Otsuka, Keisuke Matsumoto, Hitoshi Ozawa, Hirotaka Ishii

    Journal of Nippon Medical School   90 ( 2 )   138 - 140   2023.4

     More details

    Authorship:Corresponding author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:Medical Association of Nippon Medical School  

    DOI: 10.1272/jnms.jnms.2023_90-209

    researchmap

  • In situ hybridization法の高感度化手法とその神経科学未解明領域への応用 Invited Reviewed

    心平 肥後, 萌子 金谷, 友喜 水野, 一史 小澤, 篤裕 坂本, 寛高 石井

    Nihon Ika Daigaku Igakkai Zasshi   19 ( 2 )   84 - 89   2023.4

     More details

    Authorship:Last author   Language:Japanese   Publishing type:Research paper (scientific journal)   Publisher:Medical Association of Nippon Medical School  

    DOI: 10.1272/manms.19.84

    researchmap

  • Short-term depletion of plasma estrogen affects hypothalamic kisspeptin-neurokinin B-dynorphin A neurons, gonadotrophs, and pulsatile luteinizing hormone secretion in female rats Reviewed

    Kinuyo Iwata, Risako Ogata, Marimo Sato, Fuko Matsuda, Hirotaka Ishii, Hitoshi Ozawa

    Peptides   160   170929 - 170929   2023.2

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:Elsevier BV  

    DOI: 10.1016/j.peptides.2022.170929

    researchmap

  • Long-term effects of prenatal undernutrition on female rat hypothalamic KNDy neurons Reviewed

    Shiori Minabe, Kinuyo Iwata, Youki Watanabe, Hirotaka Ishii, Hitoshi Ozawa

    Endocrine Connections   12 ( 1 )   2023.1

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:Bioscientifica  

    The nutritional environment during development periods induces metabolic programming, leading to metabolic disorders and detrimental influences on human reproductive health. This study aimed to determine the long-term adverse effect of intrauterine malnutrition on the reproductive center kisspeptin-neurokinin B-dynorphin A (KNDy) neurons in the hypothalamic arcuate nucleus (ARC) of female offspring. Twelve pregnant rats were divided into ad-lib-fed (control, n  = 6) and 50% undernutrition (UN, n  = 6) groups. The UN group was restricted to 50% daily food intake of the control dams from gestation day 9 until term delivery. Differences between the two groups in terms of various maternal parameters, including body weight (BW), pregnancy duration, and litter size, as well as birth weight, puberty onset, estrous cyclicity, pulsatile luteinizing hormone (LH) secretion, and hypothalamic gene expression of offspring, were determined. Female offspring of UN dams exhibited low BW from birth to 3 weeks, whereas UN offspring showed signs of precocious puberty; hypothalamic Tac3 (a neurokinin B gene) expression was increased in prepubertal UN offspring, and the BW at the virginal opening was lower in UN offspring than that in the control group. Interestingly, the UN offspring showed significant decreases in the number of KNDy gene-expressing cells after 29 weeks of age, but the number of ARC kisspeptin-immunoreactive cells, pulsatile LH secretions, and estrous cyclicity were comparable between the groups. In conclusion, intrauterine undernutrition induced various changes in KNDy gene expression depending on the life stage. Thus, intrauterine undernutrition affected hypothalamic developmental programming in female rats.

    DOI: 10.1530/ec-22-0307

    PubMed

    researchmap

    Other Link: https://ec.bioscientifica.com/downloadpdf/journals/ec/12/1/EC-22-0307.xml

  • Optimized Mouse-on-mouse Immunohistochemical Detection of Mouse ESR2 Proteins with PPZ0506 Monoclonal Antibody Reviewed

    Mina Ozawa, Yujiro Hattori, Shimpei Higo, Mai Otsuka, Keisuke Matsumoto, Hitoshi Ozawa, Hirotaka Ishii

    ACTA HISTOCHEMICA ET CYTOCHEMICA   55 ( 5 )   159 - 168   2022.10

     More details

    Authorship:Last author, Corresponding author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:Japan Society of Histochemistry & Cytochemistry  

    Despite the physiological significance of ESR2, a lack of well-validated detection systems for ESR2 proteins has hindered progress in ESR2 research. Thus, recent identification of a specific anti-human ESR2 monoclonal antibody (PPZ0506) and its specific cross-reactivity against mouse and rat ESR2 proteins heightened momenta toward development of appropriate immunohistochemical detection systems for rodent ESR2 proteins. Building upon our previous optimization of ESR2 immunohistochemical detection in rats using PPZ0506, in this study, we further aimed to optimize mouse-on-mouse immunohistochemical detection using PPZ0506. Our assessment of several staining conditions using paraffin-embedded ovary sections revealed that intense heat-induced antigen retrieval, appropriate blocking, and appropriate antibody dilutions were necessary for optimization of mouse-on-mouse immunohistochemistry. Subsequently, we applied the optimized immunostaining method to determine expression profiles of mouse ESR2 proteins in peripheral tissues and brain subregions. Our analyses revealed more localized distribution of mouse ESR2 proteins than previously assumed. Moreover, comparison of these results with those obtained in humans and rats using PPZ0506 revealed interspecies differences in ESR2 expression. We expect that our optimized methodology for immunohistochemical staining of mouse ESR2 proteins will help researchers to solve multiple lines of controversial evidence concerning ESR2 expression.

    DOI: 10.1267/ahc.22-00043

    PubMed

    researchmap

  • Identification of Novel C-Terminally Truncated Estrogen Receptor β Variant Transcripts and Their Distribution in Humans Reviewed

    Hirotaka Ishii, Yujiro Hattori, Hitoshi Ozawa

    Journal of Nippon Medical School   88 ( 1 )   54 - 62   2021.2

     More details

    Authorship:Lead author, Corresponding author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:Medical Association of Nippon Medical School  

    BACKGROUND: The nuclear receptor genes, including estrogen receptor β (ERβ), contain non-conventional internal and terminal exons, and alternative choice of the exons yields multiple mRNA and protein variants with unique structures and functions. However, the genomic structure of the intronic and 3'-downstream regions of the human ERβ gene and the presence of novel ERβ variants with non-conventional sequences have not been re-examined in about 20 years. Therefore, we attempted to re-characterize the structure of the human ERβ gene and identify novel non-conventional exons and distinct splice variants. METHODS: Rapid amplification of cDNA 3'-end and RT-PCR cloning were used to isolate human ERβ mRNA variants from the testis. The identified cDNA sequences were mapped on the human genome assembly. Expression profiles of the variants were assessed by RT-PCR analysis. RESULTS: We cloned multiple ERβ mRNA variants with novel nucleotide sequences from the testis and identified several alternative splice sites, 3'-elongation of conventional coding exons, and novel terminal exons in the human ERβ gene. The variants encode C-terminally truncated ERβ proteins termed ERβ6, ERβ7, ERβEx. 4L, and ERβEx. 6L. Furthermore, we identified exon 7-defective forms of ERβ2/βcx, ERβ4, ERβ6, and ERβ7. Subsequently, we noted distinct expression patterns of the variants in human peripheral organs and brain subregions. CONCLUSION: This study clarified complicated genomic organization and splicing patterns of the human ERβ gene that contribute to the distinct heterogeneity of human ERβ mRNAs and proteins.

    DOI: 10.1272/jnms.jnms.2021_88-105

    PubMed

    researchmap

  • Optimization of immunohistochemical detection of rat ESR2 proteins with well-validated monoclonal antibody PPZ0506. Reviewed International journal

    Yujiro Hattori, Hirotaka Ishii, Shimpei Higo, Mai Otsuka, Moeko Kanaya, Keisuke Matsumoto, Mina Ozawa, Hitoshi Ozawa

    Molecular and cellular endocrinology   523   111145 - 111145   2021.1

     More details

    Authorship:Corresponding author   Language:English   Publishing type:Research paper (scientific journal)  

    Although there are few well-validated antibodies against ESR2 proteins, a recent validation assessment identified a specific monoclonal antibody against human ESR2 proteins (PPZ0506). Furthermore, our previous study confirmed its cross-reactivity and specificity against rodent ESR2 proteins, enabling the determination of true ESR2 distribution profiles in rodents. Therefore, we aimed to determine optimal conditions for ESR2 detection by PPZ0506 immunostaining and analyze ESR2 distribution in rats. We evaluated several staining conditions using paraffin-embedded and frozen ovary sections. Immunohistochemical staining with PPZ0506 antibody required strong antigen retrieval and appropriate antibody dilution. Subsequent immunohistochemical analysis in multiple tissues under optimized conditions revealed that rat ESR2 proteins are expressed in a more localized manner than previously assumed. Our results suggest that previous immunohistochemical studies using inadequately validated antibodies against ESR2 proteins overestimated their distribution profiles. We expect that optimized immunohistochemical detection with PPZ0506 antibody can help researchers solve several conflicting problems in ESR2 research. (150 words).

    DOI: 10.1016/j.mce.2020.111145

    PubMed

    researchmap

  • Quantitative expression data of human estrogen receptor α variants in non-functioning pituitary adenomas obtained by reverse transcription-digital polymerase chain reaction analysis Reviewed International journal

    Yujiro Hattori, Hirotaka Ishii, Shigeyuki Tahara, Akio Morita, Hitoshi Ozawa

    Data in Brief   33   106452 - 106452   2020.12

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:Elsevier BV  

    Expression profiles of gonadal steroid receptor variants have been reportedly associated with malignancy in breast and prostate cancers [1,2]. However, such associations with pituitary tumors remain unclear. Therefore, the expression levels of the wild-type ESR1 (ERα66) and the ESR1 variants (ERαi34, ERαi45c, and ERαΔ5) transcripts encoding constitutively active ERα proteins with C-terminal truncation in non-functioning pituitary adenomas (NFPAs) were evaluated using reverse transcription-digital polymerase chain reaction. The results revealed that the expression levels of the variants were approximately two orders of magnitude lower than that of ERα66 in NFPAs. These data were based on our previous article entitled "Accurate assessment of estrogen receptor profiles in non-functioning pituitary adenomas using RT-digital PCR and immunohistochemistry" [3].

    DOI: 10.1016/j.dib.2020.106452

    PubMed

    researchmap

  • Accurate assessment of estrogen receptor profiles in non-functioning pituitary adenomas using RT-digital PCR and immunohistochemistry. Reviewed International journal

    Yujiro Hattori, Hirotaka Ishii, Shigeyuki Tahara, Akio Morita, Hitoshi Ozawa

    Life sciences   260   118416 - 118416   2020.9

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    BACKGROUND: Non-functioning pituitary adenomas (NFPAs) are common pituitary tumors, and surgery is generally the only treatment option. Few attempts have been made to explore target molecules for the development of NFPA pharmacological treatments. METHOD: We quantitatively assessed the expression profiles of estrogen receptor (ER) transcripts and proteins in NFPA samples, using reverse transcription-digital polymerase chain reaction (RT-dPCR) and immunohistochemistry, and further investigated the correlations between the expression levels of ER and those of downstream responsive genes. All patients had undergone surgery at the same high-volume hospital. A total of 20 patients with NFPAs were included. All patients were new-onset, and none were diagnosed with intratumoral hemorrhages or cysts. RESULTS: NFPA samples exhibited a bimodal ESR1 expression pattern and were categorized into significantly different high- and low-ESR1 expression level groups (P < 0.05). In contrast, expression levels of ESR1 variants and ESR2 could barely be detected. Similar results were obtained through the immunohistochemical staining of NFPAs, using well-validated antibodies against ERs. The expression levels of ESR1 positively correlated with those of GREB1, an estrogen-responsive gene [correlation coefficient (r) = 0.623, P = 0.003]. CONCLUSIONS: ESR1 expression levels in NFPAs exhibited a bimodal pattern and were positively correlated with GREB1 expression levels. The accurate assessment of ER expression levels may further advance future NFPA-related research.

    DOI: 10.1016/j.lfs.2020.118416

    PubMed

    researchmap

  • GnRH(1-5), a metabolite of gonadotropin-releasing hormone, enhances luteinizing hormone release via activation of kisspeptin neurons in female rats. Reviewed

    Nahoko Ieda, Assadullah, Shiori Minabe, Kana Ikegami, Youki Watanabe, Yusuke Sugimoto, Arisa Sugimoto, Narumi Kawai, Hirotaka Ishii, Naoko Inoue, Yoshihisa Uenoyama, Hiroko Tsukamura

    Endocrine journal   67 ( 4 )   409 - 418   2020.4

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Accumulating evidence suggests that kisspeptin neurons in the arcuate nucleus (ARC), which coexpress neurokinin B and dynorphin, are involved in gonadotropin-releasing hormone (GnRH)/luteinizing hormone (LH) pulse generation, while the anteroventral periventricular nucleus (AVPV) kisspeptin neurons are responsible for GnRH/LH surge generation. The present study aims to examine whether GnRH(1-5), a GnRH metabolite, regulates LH release via kisspeptin neurons. GnRH(1-5) was intracerebroventricularly injected to ovariectomized and estrogen-treated Wistar-Imamichi female rats. Immediately after the central GnRH(1-5) administration at 2 nmol, plasma LH concentration increased, resulting in significantly higher levels of the area under the curve and baseline of plasma LH concentrations compared to vehicle-injected controls. On the other hand, in Kiss1 knockout rats, GnRH(1-5) administration failed to affect LH secretion, suggesting that the facilitatory effect of GnRH(1-5) on LH release is mediated by kisspeptin neurons. Double in situ hybridization (ISH) for Kiss1 and Gpr101, a GnRH(1-5) receptor gene, revealed that few Kiss1-expressing cells coexpress Gpr101 in both ARC and AVPV. On the other hand, double ISH for Gpr101 and Slc17a6, a glutamatergic marker gene, revealed that 29.2% of ARC Gpr101-expressing cells coexpress Slc17a6. Further, most of the AVPV and ARC Kiss1-expressing cells coexpress Grin1, a gene encoding a subunit of NMDA receptor. Taken together, these results suggest that the GnRH(1-5)-GPR101 signaling facilitates LH release via indirect activation of kisspeptin neurons and that glutamatergic neurons may mediate the signaling. This provides a new aspect of kisspeptin- and GnRH-neuronal communication with the presence of stimulation from GnRH to kisspeptin neurons in female rats.

    DOI: 10.1507/endocrj.EJ19-0444

    PubMed

    researchmap

  • Identification of a novel C-terminally truncated estrogen receptor α variant (ERαi34) with constitutive transactivation and estrogen receptor antagonist resistance. Reviewed International journal

    Hirotaka Ishii, Yujiro Hattori, Hitoshi Ozawa

    Molecular and cellular endocrinology   503   110693 - 110693   2020.3

     More details

    Authorship:Lead author, Corresponding author   Language:English   Publishing type:Research paper (scientific journal)  

    Constitutively active estrogen receptor α (ERα) variants with C-terminal truncation are candidate molecules for gain of both endocrine- and chemotherapy-resistance in estrogen-sensitive tumors. Our previous reports using artificially truncated ERα constructs demonstrated that ERα variants encoded in 1-2-3-cryptic exon- and 1-2-3-4-cryptic exon-types of transcripts have potentials to display constitutive transactivation of an estrogen response element reporter. However, naturally occurring 1-2-3-cryptic exon-type ERα variants have not been cloned yet. Therefore, the present study was designed to identify naturally occurring ERα variants encoded in 1-2-3-cryptic exon-type ERα transcripts. We cloned a novel C-terminally truncated ERα variant (ERαi34) encoded in a 1-2-3-i34 transcript from MCF-7 cells and characterized its constitutive and ER antagonist-resistant transactivation in transfected cells. Stable transfection of the variant into MCF-7 cells augmented basal cell proliferation insensitive to fulvestrant. Collectively, we validated the structure-based mechanisms underlying constitutive and ER antagonist-resistant transactivation by C-terminally truncated ERα variants.

    DOI: 10.1016/j.mce.2019.110693

    PubMed

    researchmap

  • Applicability of Anti-Human Estrogen Receptor β Antibody PPZ0506 for the Immunodetection of Rodent Estrogen Receptor β Proteins. Reviewed International journal

    Hirotaka Ishii, Mai Otsuka, Moeko Kanaya, Shimpei Higo, Yujiro Hattori, Hitoshi Ozawa

    International journal of molecular sciences   20 ( 24 )   2019.12

     More details

    Authorship:Lead author, Corresponding author   Language:English   Publishing type:Research paper (scientific journal)  

    Several lines of controversial evidence concerning estrogen receptor β (ERβ) remain to be solved because of the unavailability of specific antibodies against ERβ. The recent validation analysis identified a monoclonal antibody (PPZ0506) with sufficient specificity against human ERβ. However, the specificity and cross-reactivity of PPZ0506 antibody against ERβ proteins from laboratory animals have not been confirmed. In the present study, we aimed to validate the applicability of PPZ0506 to rodent studies. The antibody exhibited specific cross-reactivity against mouse and rat ERβ proteins in immunoblot and immunocytochemical experiments using transfected cells. In immunohistochemistry for rat tissue sections, PPZ0506 showed immunoreactive signals in the ovary, prostate, and brain. These immunohistochemical profiles of rat ERβ proteins in rat tissues accord well with its mRNA expression patterns. Although the antibody was reported to show the moderate signals in human testis, no immunoreactive signals were observed in rat testis. Subsequent RT-PCR analysis revealed that this species difference in ERβ expression resulted from different expression profiles related to the alternative promoter usage between humans and rats. In conclusion, we confirmed applicability of PPZ0506 for rodent ERβ studies, and our results provide a fundamental basis for further examination of ERβ functions.

    DOI: 10.3390/ijms20246312

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0003-2044-316X

  • Stable Reference Gene Selection for Reverse Transcription-Quantitative PCR (RT-qPCR) Analyses in Orthopaedic Research Reviewed

    Hiroshi Watanabe, Hirotaka Ishii, Yasushi Oshima, Shinro Takai, Hitoshi Ozawa

    日本医科大学医学会雑誌   15 ( 1 )   24 - 31   2019.2

     More details

    Authorship:Corresponding author   Language:Japanese  

    DOI: 10.1272/manms.15.24

    researchmap

  • Genomic Organization of the 5'-untranslated Regions of Estrogen Receptor α Genes Invited Reviewed

    石井 寛高, 服部 裕次郎, 小澤 一史

    日本医科大学医学会雑誌   14 ( 4 )   157 - 164   2018.10

     More details

    Authorship:Lead author, Corresponding author   Language:Japanese   Publisher:日本医科大学医学会  

    researchmap

  • Suitable reference gene selection for gene expression studies in knee osteoarthritis synovium using quantitative PCR analysis. Reviewed International journal

    Hiroshi Watanabe, Hirotaka Ishii, Kenji Takahashi, Shinro Takai, Hitoshi Ozawa

    Connective tissue research   59 ( 4 )   356 - 368   2018.7

     More details

    Authorship:Corresponding author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:TAYLOR & FRANCIS INC  

    Osteoarthritis (OA) is the leading cause of musculoskeletal disability in the elderly. Insights into the biological features of OA are obtained by characterization of the molecular features by gene expression profiling using reverse transcription-quantitative PCR (RT-qPCR). However, it has recently become evident that the use of suitable reference genes is required for appropriate normalization of this technique. Here total RNA was isolated from the synovium of 18 men and 20 women who underwent total knee arthroplasty for knee OA (KOA). We validated the expression stability of 7 candidate housekeeping genes (ACTB, B2M, GAPDH, HPRT1, RPL13A, SDHA, and YWHAZ) in the synovium of KOA with 3 commonly used algorithms (geNorm, NormFinder, and BestKeeper). Additionally, we evaluated expression profiles of the steroid hormone receptor (AR, ESR1, ESR2, GR, MR, and PR) and proinflammatory cytokines (IL1B and IL6) genes in the synovium and their correlations with the risk factors of KOA, using the most and least stable housekeeping genes for comparison. Results showed that HPRT1 was the most stable gene, whereas B2M was the least stable. RT-qPCR analysis revealed sexually dimorphic expression of AR, IL1B, and IL6; intercorrelations between steroid hormone receptor expression levels and female-specific correlations of IL1B expression with ESR1 and PR expression, IL6 expression with ESR1 and GR expression, and body mass index with AR and PR expression; and the choice of the least stable reference gene altered several correlations and statistical significances. In conclusion, HPRT1 was identified as the suitable reference gene for normalization in the OA synovium.

    DOI: 10.1080/03008207.2017.1391234

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0003-2044-316X

  • Morphological Analysis of Trafficking and Processing of Anionic and Cationic Liposomes in Cultured Cells. Reviewed

    Yuji Tomori, Norio Iijima, Shuji Hinuma, Hirotaka Ishii, Ken Takumi, Shinro Takai, Hitoshi Ozawa

    Acta histochemica et cytochemica   51 ( 2 )   81 - 92   2018.4

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:JAPAN SOC HISTOCHEMISTRY & CYTOCHEMISTRY  

    Liposomes, artificial phospholipid vesicles, have been developed as a non-viral drug delivery system to allow contained agents to be efficiently delivered to target sites via systemic circulation. Liposomes have been used as a gene transfer tool with cultured cells; however, their precise trafficking and processing remain uncertain. Furthermore, liposomes with different surface charges are known to exhibit distinct properties. The purpose of the current study was to elucidate the intracellular trafficking and processing of liposomes with anionic and cationic surface charges from a morphological view point. We found that cationic liposomes (CLs) were more effectively taken by the cells than anionic liposomes (ALs). Confocal laser scanning microscopy and transmission electron microscopy demonstrated distinct intracellular localization and processing patterns of ALs and CLs. ALs and their contents were localized in lysosomes but not in cytosol, indicating that ALs are subjected to the endosome-lysosome system. In contrast, contents of CLs were distributed mainly in the cytosol. CLs appear to disturb the cell membrane and then collapse to release their contents into the cytosol. It is feasible that the contents of CLs enter the cytosol directly rather than via the endosome-lysosome system.

    DOI: 10.1267/ahc.17021

    Web of Science

    PubMed

    researchmap

  • Co-expression of the calcitonin receptor gene in the hypothalamic kisspeptin neurons in female rats. Reviewed

    Assadullah, Nahoko Ieda, Narumi Kawai, Hirotaka Ishii, Kunio Ihara, Naoko Inoue, Yoshihisa Uenoyama, Hiroko Tsukamura

    Reproductive medicine and biology   17 ( 2 )   164 - 172   2018.4

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:Wiley-Blackwell  

    Purpose: Hypothalamic kisspeptin neurons are considered to play a critical role in regulating mammalian reproduction and integrating humoral and neuronal inputs that control gonadotropin-releasing hormone (GnRH)/gonadotropin release. The present study aimed to investigate the upstream regulator candidates for kisspeptin neurons. Methods: Visualized kisspeptin neurons that were taken from the arcuate nucleus (ARC) of Kiss1-tdTomato rats were subjected to next-generation sequencing (NGS) analysis. In situ hybridization (ISH) for the calcitonin receptor gene (Calcr) was performed throughout the whole forebrain of ovariectomized wild-type female rats that had been implanted with a negative feedback level of estrogen, because the Calcr expression was evident in the ARC kisspeptin neurons from the NGS analysis. Then, a double ISH was performed for the Calcr and kisspeptin gene (Kiss1) in the brain regions, containing either the anteroventral periventricular nucleus (AVPV) or ARC of the female rats. Results: The NGS analysis revealed that the Calcr was highly expressed in the ARC kisspeptin neurons. It was found that the Calcr was co-expressed in 12% and 22% of the Kiss1-expressing cells in the ARC and AVPV, respectively. Conclusion: The present study suggests that calcitonin receptor signaling could be involved in the regulation of reproductive function through the direct control of the ARC and/or AVPV kisspeptin neurons, and then GnRH/gonadotropin release.

    DOI: 10.1002/rmb2.12085

    PubMed

    researchmap

  • Characterization of rodent constitutively active estrogen receptor α variants and their constitutive transactivation mechanisms. Reviewed International journal

    Hirotaka Ishii, Yujiro Hattori, Arisa Munetomo, Hiroshi Watanabe, Yasuo Sakuma, Hitoshi Ozawa

    General and comparative endocrinology   248   16 - 26   2017.7

     More details

    Authorship:Lead author, Corresponding author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:ACADEMIC PRESS INC ELSEVIER SCIENCE  

    Estrogen receptor α (ERα) mRNAs exhibit remarkable heterogeneity owing to complicated alternative splicing. Some encode C-terminally-truncated ERα proteins, which display ligand-independent transactivation or dominant-negative activity. We previously characterized C-terminally-truncated ERα mRNA variants with cryptic sequences in humans and mice, and demonstrated that helices in the ligand-binding domains (LBDs) of ERα variants contribute to ligand-independent transcriptional activity. However, existence of non-conventional coding exons and generation of constitutively active ERα variants have remained to be examined in rats. To comparatively analyze modular organization and splicing profiles of the ERα genes, the range of C-terminally-truncated ERα variants was explored in rats and mice using rapid amplification of cDNA ends and RT-PCR cloning. Furthermore, their functions were determined in transiently transfected cells using expression constructs and luciferase reporter assays. Multiple cryptic exons and C-terminally-truncated ERα variant mRNAs were identified in rats and mice. Naturally occurring and artificially truncated variants/constructs lacking helix 5 potentially exhibited gain-of-function in transfected cells. Although cryptic exons and splicing profiles were poorly conserved among humans, mice, and rats, constitutively active variants were generated from the ERα genes. Moreover, the primary mechanism of ligand-independent activation by C-terminally-truncated ERα variants is C-terminus to helix 5 deletion in the LBD. This comparative study documented the complexity of ERα genes, mRNAs, and proteins, and further determined the underlying structural basis of ligand-independent activation by C-terminally-truncated ERα variants.

    DOI: 10.1016/j.ygcen.2017.04.009

    Web of Science

    PubMed

    researchmap

  • Subunit profiling and functional characteristics of acetylcholine receptors in GT1-7 cells. Reviewed

    Yuki Arai, Hirotaka Ishii, Makito Kobayashi, Hitoshi Ozawa

    The journal of physiological sciences : JPS   67 ( 2 )   313 - 323   2017.3

     More details

    Authorship:Corresponding author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:SPRINGER JAPAN KK  

    GnRH neurons form a final common pathway for the central regulation of reproduction. Although the involvement of acetylcholine in GnRH secretion has been reported, direct effects of acetylcholine and expression profiles of acetylcholine receptors (AChRs) still remain to be studied. Using immortalized GnRH neurons (GT1-7 cells), we analyzed molecular expression and functionality of AChRs. Expression of the mRNAs were identified in the order α7 > β2 = β1 ≧ α4 ≧ α5 = β4 = δ > α3 for nicotinic acetylcholine receptor (nAChR) subunits and m4 > m2 for muscarinic acetylcholine receptor (mAChR) subtypes. Furthermore, this study revealed that α7 nAChRs contributed to Ca2+ influx and GnRH release and that m2 and m4 mAChRs inhibited forskolin-induced cAMP production and isobutylmethylxanthine-induced GnRH secretion. These findings demonstrate the molecular profiles of AChRs, which directly contribute to GnRH secretion in GT1-7 cells, and provide one possible regulatory action of acetylcholine in GnRH neurons.

    DOI: 10.1007/s12576-016-0464-1

    Web of Science

    PubMed

    researchmap

  • Characterization of sevoflurane effects on Per2 expression using ex vivo bioluminescence imaging of the suprachiasmatic nucleus in transgenic rats. Reviewed International journal

    Izumi Matsuo, Norio Iijima, Ken Takumi, Shimpei Higo, Satoko Aikawa, Megumi Anzai, Hirotaka Ishii, Atsuhiro Sakamoto, Hitoshi Ozawa

    Neuroscience research   107   30 - 7   2016.6

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:ELSEVIER IRELAND LTD  

    The inhalation anesthetic sevoflurane suppresses Per2 expression in the suprachiasmatic nucleus (SCN) in rodents. Here, we investigated the intra-SCN regional specificity, time-dependency, and pharmacological basis of sevoflurane-effects. Bioluminescence image was taken from the SCN explants of mPer2 promoter-destabilized luciferase transgenic rats, and each small regions of interest (ROI) of the image was analyzed. Sevoflurane suppressed bioluminescence in all ROIs, suggesting that all regions in the SCN are sensitive to sevoflurane. Clear time-dependency in sevoflurane effects were also observed; application during the trough phase of the bioluminescence cycle suppressed the subsequent increase in bioluminescence and resulted in a phase delay of the cycle; sevoflurane applied during the middle of the ascending phase induced a phase advance; sevoflurane on the descending phase showed no effect. These results indicate that the sevoflurane effect may depend on the intrinsic state of circadian machinery. Finally, we examined the involvement of GABAergic signal transduction in the sevoflurane effect. Co-application of both GABAA and GABAB receptor antagonists completely blocked the effect of sevoflurane on the bioluminescence rhythm, suggesting that sevoflurane inhibits Per2 expression via GABAergic signal transduction. Current study elucidated the anesthetic effects on the molecular mechanisms of circadian rhythm.

    DOI: 10.1016/j.neures.2015.11.010

    Web of Science

    PubMed

    researchmap

  • Establishment of an in vitro cell line experimental system for the study of inhalational anesthetic mechanisms. Reviewed International journal

    Seiji Nagamoto, Norio Iijima, Hirotaka Ishii, Ken Takumi, Shimpei Higo, Satoko Aikawa, Megumi Anzai, Izumi Matsuo, Shinji Nakagawa, Naoyuki Takashima, Yasufumi Shigeyoshi, Atsuhiro Sakamoto, Hitoshi Ozawa

    Neuroscience letters   620   163 - 8   2016.5

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:ELSEVIER IRELAND LTD  

    General anesthesia affects the expression of clock genes in various organs. Expression of Per2, a core component of the circadian clock, is markedly and reversibly suppressed by sevoflurane in the suprachiasmatic nucleus (SCN), and is considered to be a biochemical marker of anesthetic effect in the brain. The SCN contains various types of neurons, and this complexity makes it difficult to investigate the molecular mechanisms of anesthesia. Here, we established an in vitro experimental system using a cell line to investigate the mechanisms underlying anesthetic action. Development of the system comprised two steps: first, we developed a system for application of inhalational anesthetics and incubation; next, we established cultures of anesthetic-responsive cells expressing mPer2 promoter-dLuc. GT1-7 cells, derived from the mouse hypothalamus, responded to sevoflurane by reversibly decreasing mPer2-promoter-driven bioluminescence. Interestingly, the suppression of bioluminescence was found only in the serum-starved GT1-7 cells, which showed neuron-like morphology, but not in growing cells, suggesting that neuron-like characteristics are required for anesthetic effects in GT1-7 cells.

    DOI: 10.1016/j.neulet.2016.04.005

    Web of Science

    PubMed

    researchmap

  • Human C-terminally truncated ERα variants resulting from the use of alternative exons in the ligand-binding domain. Reviewed International journal

    Yujiro Hattori, Hirotaka Ishii, Arisa Munetomo, Hiroshi Watanabe, Akio Morita, Yasuo Sakuma, Hitoshi Ozawa

    Molecular and cellular endocrinology   425 ( C )   111 - 22   2016.4

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:ELSEVIER IRELAND LTD  

    The nuclear receptor genes contain alternative internal and terminal exons, with alternative exon incorporation yielding mRNA variants that encode various receptor types, including some with C-terminal truncation that exhibit constitutive activation or dominant-negative transcriptional transactivation. However, C-terminally truncated estrogen receptor α (ERα) variants with alternative sequences have rarely been reported in humans. Therefore, we assessed human ERα genomic organization and alternative splicing profiles, and identified both alternative exons and C-terminally truncated ERα variants. These naturally occurring C-terminally truncated ERα proteins were localized in the nuclei of transfected cells. In addition, ERαi45c and ERαΔ5 variants exhibited constitutive transactivation of an estrogen responsive element-driven promoter in transfected cells. We manufactured expression vectors encoding artificially truncated ERα constructs and evaluated their transactivation abilities to establish mechanisms determining the constitutive activity and dominant-negative properties of truncated variants. Lack of the region encoded in exon 8 eliminated basal and ligand-induced transcriptional transactivation. The C-terminally truncated ERα variants/constructs containing the helices 5 in their ligand-binding domains did not exhibit constitutive transactivation. Furthermore, we demonstrated that truncation from C-termini to helices 5 in the variant ligand-binding domains was required for constitutive activation and found that the remnant regions of the ligand-binding domains and variant-specific sequences influenced transcriptional transactivation efficiency. In conclusion, we elucidated the structural and functional features of novel C-terminally truncated ERα variants and revealed the mechanisms underlying constitutive transactivation by C-terminally truncated nuclear receptor variants.

    DOI: 10.1016/j.mce.2016.01.026

    Web of Science

    PubMed

    researchmap

  • Association between steroid hormone receptors expression in the synovium and body mass index in human osteoarthritis Reviewed

    H. Watanabe, K. Takahashi, H. Ishii, N. Iizawa, H. Kawaji, T. Majima, H. Ozawa, S. Takai

    Osteoarthritis and Cartilage   24   S68 - S69   2016.4

     More details

    Publishing type:Research paper (scientific journal)   Publisher:Elsevier BV  

    DOI: 10.1016/j.joca.2016.01.150

    researchmap

  • Puerperal and parental experiences alter rat preferences for pup odors via changes in the oxytocin system. Reviewed

    Arisa Munetomo, Hirotaka Ishii, Takenori Miyamoto, Yasuo Sakuma, Yasuhiko Kondo

    The Journal of reproduction and development   62 ( 1 )   17 - 27   2016

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:SOCIETY REPRODUCTION & DEVELOPMENT-SRD  

    In the rat, induction of maternal behavior depends on the parity of the female. For example, nulliparous (NP) females need longer exposure to pups than multiparous (MP) or lactating (L) females to exhibit similar maternal behavior. In this study, we investigated the role of brain oxytocin in the approaching behavior of these female rats. Olfactory preferences for pup odors were examined for 8 consecutive days. Each preference test was followed by direct overnight exposure to pups. On the 8th day, MP and L, but not NP females showed robust pup-odor preferences. After the behavioral test, half of the females were exposed to pups for 2 h, whereas the other half were not. The females were then sacrificed to analyze brain oxytocin (OXT) and vasopressin (AVP) activities by cFos immunohistochemistry and to quantify their receptor mRNA expression using real-time PCR. In the paraventricular nucleus (PVN), the percentage of cFos-positive OXT neurons was significantly larger in MP and L females than in NP females after pup exposure. No significant differences were found in cFos expression in OXT neurons of the supraoptic nucleus (SON) or in AVP neurons of either the PVN or SON. Expression of OXT receptor mRNA in the medial preoptic area and amygdala of the control groups was also higher in MP females than in NP females. Finally, we demonstrated that infusion of OXT into the lateral ventricle of NP females promoted preferences for pup odors. These results indicate that puerperal and parental experiences enhance the responsiveness of OXT neurons in the PVN to pup stimuli and establish olfactory preferences for these odors in a parity-dependent manner.

    DOI: 10.1262/jrd.2015-046

    Web of Science

    PubMed

    researchmap

  • Characterization of the fundamental properties of the N-terminal truncation (Δ exon 1) variant of estrogen receptor α in the rat. Reviewed International journal

    Yujiro Hattori, Hirotaka Ishii, Akio Morita, Yasuo Sakuma, Hitoshi Ozawa

    Gene   571 ( 1 )   117 - 25   2015.10

     More details

    Authorship:Corresponding author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:ELSEVIER SCIENCE BV  

    The estrogen receptor α (ERα) directs transactivation of target genes, and splice variants have been shown to exhibit altered activation properties. We previously documented the complicated alternative promoter usage and splicing patterns of the rat ERα gene; however, the information was restricted to a few specific organs. Therefore, we re-examined the rat mRNA profiles of ERα, including the generation of the exon 1-skipping, ERα46 transcript in a wider variety of rat organs and further characterized the fundamental functional properties of rat ERα46 variants. With the use of RT-PCR, we discovered unique distribution and splicing patterns for promoter-specific ERα isoforms, as well as the extensive expression of the Δ exon 1 variant in the rat. Similar to wild-type ERα, an immunocytochemical analysis showed a predominant localization of ERα46 proteins in the nuclei of transfected cells. Luciferase reporter assays revealed that ERα46 variants stimulated the transcriptional activity of an estrogen response element-driven promoter in response to estrogen. In addition, the variants exhibited distinct transactivation and reactivity to 4-hydroxytamoxifen in different cell types. Although the alternative splicing patterns are species-specific, the profiles of the alternative use of promoters, and the fundamental properties of the rat ERα46 variant are similar to those of human and mouse homologs. Therefore, the present study provides fundamental and useful information for further research into the regulation and functions of ERα gene variants.

    DOI: 10.1016/j.gene.2015.06.086

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0003-2044-316X

  • Novel splicing events and post-transcriptional regulation of human estrogen receptor α E isoforms. Reviewed International journal

    Hirotaka Ishii, Momoko Kobayashi, Arisa Munetomo, Takenori Miyamoto, Yasuo Sakuma

    The Journal of steroid biochemistry and molecular biology   133 ( 1 )   120 - 8   2013.1

     More details

    Authorship:Lead author, Corresponding author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:PERGAMON-ELSEVIER SCIENCE LTD  

    Expression of the estrogen receptor α (ERα) gene is subject to complex regulation. To elucidate the mechanisms of this regulation, the genomic organization and the physiological role of the multiple 5'-untranslated regions (5'-UTRs) must be determined. Here, we investigated the expression and splicing patterns of the human ERα E isoforms. We identified two novel untranslated exons, N1 and N2, in the 5'-region of the human ERα gene and multiple E isoform mRNA variants generated by alternative usage of non-coding internal exons. Expression of the N1-containing variants was observed only in the human breast adenocarcinoma cell line, MCF7, while the N2-containing variants were expressed in the adult liver and MCF7 cells. We examined post-transcriptional regulation of the variant mRNAs using luciferase reporter assays and quantitative PCR. The insertion of untranslated internal exons into the 5'-UTRs of the E isoforms reduced their translation efficiency, but barely influenced mRNA turnover. Our results indicate that the genomic organization of the human ERα gene and the splicing profiles of the human ERα E isoforms are more complicated than previously reported. Furthermore, the 5'-UTRs of the E isoforms post-transcriptionally control human ERα expression mainly through translational repression.

    DOI: 10.1016/j.jsbmb.2012.09.027

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0003-2044-316X

  • Somatostatin inhibition of GnRH neuronal activity and the morphological relationship between GnRH and somatostatin neurons in rats. Reviewed International journal

    Makiko Koyama, Chengzhu Yin, Hirotaka Ishii, Yasuo Sakuma, Masakatsu Kato

    Endocrinology   153 ( 2 )   806 - 14   2012.2

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:ENDOCRINE SOC  

    In rodents, GnRH neurons are diffusely distributed from the medial septum through to the medial preoptic area and control gonadal functions through the pituitary. The activity of GnRH neurons is regulated by a variety of bioactive substances, including the inhibitory peptide somatostatin. In the present study, we focused on somatostatin because intracerebroventricular injection of somatostatin inhibits the LH surge in rats and reduces LH secretion in ewes. Somatostatin also decreases GnRH release from rat hypothalamic slices. In mice, somatostatin is also thought to suppress GnRH neuronal activity through contact on the soma of GnRH neurons. However, similar data are missing in rats. Moreover, rat GnRH neurons receive only a few synaptic inputs. In this study, we assessed the morphological relationship between GnRH and somatostatin neurons. Confocal microscopy on the sections from the medial septum through medial preoptic area revealed about 35 close contacts per rat between the GnRH and somatostatin neuronal fibers in the organum vasculosum of the lamina terminalis region. No contact of somatostatin fibers on the GnRH neuronal somata was observed. Multicell RT-PCR for somatostatin receptor mRNA in rat GnRH neurons was also performed, which revealed moderate expression of somatostatin receptor subtypes 1-5. In addition, patch clamp experiments were carried out in acute slice preparations. Somatostatin suppressed neuronal firing in cells recorded in a cell-attached configuration and also induced whole-cell outward currents in GnRH neurons. These findings suggest that somatostatin directly inhibits the activity of rat GnRH neurons through volume transmission in the organum vasculosum of the lamina terminalis region.

    DOI: 10.1210/en.2011-1374

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0003-2044-316X

  • Complex organization of the 5'-untranslated region of the mouse estrogen receptor α gene: identification of numerous mRNA transcripts with distinct 5'-ends. Reviewed International journal

    Hirotaka Ishii, Yasuo Sakuma

    The Journal of steroid biochemistry and molecular biology   125 ( 3-5 )   211 - 8   2011.7

     More details

    Authorship:Lead author, Corresponding author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:PERGAMON-ELSEVIER SCIENCE LTD  

    The 5'-untranslated region (5'-UTR) of the estrogen receptor α (ERα) gene plays an important role in determining its tissue-specific expression. We examined the 5'-UTRs of the mouse ERα mRNA variants in depth using the Basic Local Alignment Search Tool (BLAST), rapid amplification of 5'-cDNA ends (5'-RACE) and RT-PCR. We demonstrated the presence of multiple variants containing unique 5'-UTRs. We mapped the cDNA sequences onto the mouse genome, and found that both alternative splicing from four different leader exons (A, C, F1, and H) to exon 1, and combinations of 12 internal exons (X1, X2, X3, X4, F2/X5, X6, X7, X8, X9, X10, X11, and B) generate multiple ERα transcripts. Mouse exon B, that has homologies with human exon B and rat exon 0T, was used as an internal exon, not as a leader exon. RT-PCR analysis revealed distinct expression patterns of the variants, suggesting that the alternative promoter usage and alternative splicing are regulated in a tissue-specific manner. Our results indicate that the genomic organization of the mouse ERα gene is complicated as previously shown in the rat ERα gene. In addition, both alternative promoter usage and alternative splicing contribute to the remarkable mRNA diversity of the mouse ERα gene.

    DOI: 10.1016/j.jsbmb.2011.03.004

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0003-2044-316X

  • Identification of C-terminally and N-terminally truncated estrogen receptor α variants in the mouse. Reviewed International journal

    Hirotaka Ishii, Yuri Shoda, Kentaro Yomogida, Tomohiro Hamada, Yasuo Sakuma

    The Journal of steroid biochemistry and molecular biology   124 ( 1-2 )   38 - 46   2011.3

     More details

    Authorship:Lead author, Corresponding author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:PERGAMON-ELSEVIER SCIENCE LTD  

    We re-examined mouse ERα mRNA variants using rapid amplification of cDNA ends (RACE) and RT-PCR. Our analysis showed the presence of several mRNA variants containing unique 5'- or 3'-nucleotide sequences. We mapped the cDNA sequences on the mouse genome, and identified four novel 3'-terminal and 5'-leader exons in the intronic region between exons 4 and 5. RT-PCR analysis revealed that the expression patterns of the C-terminally truncated ERα products (CTERPs) were similar to that of Wild-type ERα and that the N-terminally truncated ERα products (NTERPs) appeared to have different expression profiles. Moreover, we constructed expression vectors and analyzed the subcellular localization and the transcriptional activation abilities of the variant proteins in transfected HEK293 cells using immunocytochemistry and luciferase reporter assay. The CTERP variants localized in the nuclei and constitutively activated estrogen response element (ERE)-driven promoters, while the NTERP variant was located in the extra-nuclear regions and had no ability to activate the ERE promoters in the presence or absence of 10 nM estradiol. Our results indicate that the mouse ERα gene is more complex than previously thought in terms of genomic organization and that alternative splicing and alternative usage of intronic promoters contribute to the remarkable diversity of ERα mRNAs and proteins.

    DOI: 10.1016/j.jsbmb.2011.01.003

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0003-2044-316X

  • Identification of novel splicing events and post-transcriptional regulation of human estrogen receptor α F isoforms. Reviewed International journal

    Momoko Kobayashi, Hirotaka Ishii, Yasuo Sakuma

    Molecular and cellular endocrinology   333 ( 1 )   55 - 61   2011.2

     More details

    Authorship:Corresponding author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:ELSEVIER IRELAND LTD  

    The 5'-untranslated regions (5'-UTRs) of the estrogen receptor α (ERα) mRNAs play important roles in the modulation of translation. To elucidate the mechanisms regulating human ERα gene expression, it is necessary to determine its genomic organization and the roles of the multiple 5'-UTRs. We therefore examined the splicing and expression profiles of the human ERα F isoforms. A novel non-coding exon E3 was found in the 5'-region of the human ERα gene and six F isoform mRNA variants were identified. The variants revealed the preferred expression patterns. Post-transcriptional regulation was also investigated. The 5'-UTRs of the F isoforms decreased the translation efficiency but had no effect on mRNA stability. The results indicate that the organization and splicing patterns of the human ERα gene are more complex than previously reported, and that the 5'-UTRs of the F isoforms contribute to the post-transcriptional control of human ERα gene expression through translation repression.

    DOI: 10.1016/j.mce.2010.12.003

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0003-2044-316X

  • 中枢神経における性差科学の現状

    濱田 知宏, 石井 寛高, 佐久間 康夫

    ファルマシア   47 ( 3 )   213 - 217   2011

     More details

    Language:Japanese   Publisher:公益社団法人 日本薬学会  

    DOI: 10.14894/faruawpsj.47.3_213

    CiNii Books

    researchmap

  • Endogenous synthesis of corticosteroids in the hippocampus. Reviewed International journal

    Shimpei Higo, Yasushi Hojo, Hirotaka Ishii, Yoshimasa Komatsuzaki, Yuuki Ooishi, Gen Murakami, Hideo Mukai, Takeshi Yamazaki, Daiichiro Nakahara, Anna Barron, Tetsuya Kimoto, Suguru Kawato

    PloS one   6 ( 7 )   e21631   2011

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:PUBLIC LIBRARY SCIENCE  

    BACKGROUND: Brain synthesis of steroids including sex-steroids is attracting much attention. The endogenous synthesis of corticosteroids in the hippocampus, however, has been doubted because of the inability to detect deoxycorticosterone (DOC) synthase, cytochrome P450(c21). METHODOLOGY/PRINCIPAL FINDINGS: The expression of P450(c21) was demonstrated using mRNA analysis and immmunogold electron microscopic analysis in the adult male rat hippocampus. DOC production from progesterone (PROG) was demonstrated by metabolism analysis of (3)H-steroids. All the enzymes required for corticosteroid synthesis including P450(c21), P450(2D4), P450(11β1) and 3β-hydroxysteroid dehydrogenase (3β-HSD) were localized in the hippocampal principal neurons as shown via in situ hybridization and immunoelectron microscopic analysis. Accurate corticosteroid concentrations in rat hippocampus were determined by liquid chromatography-tandem mass spectrometry. In adrenalectomized rats, net hippocampus-synthesized corticosterone (CORT) and DOC were determined to 6.9 and 5.8 nM, respectively. Enhanced spinogenesis was observed in the hippocampus following application of low nanomolar (10 nM) doses of CORT for 1 h. CONCLUSIONS/SIGNIFICANCE: These results imply the complete pathway of corticosteroid synthesis of 'pregnenolone →PROG→DOC→CORT' in the hippocampal neurons. Both P450(c21) and P450(2D4) can catalyze conversion of PROG to DOC. The low nanomolar level of CORT synthesized in hippocampal neurons may play a role in modulation of synaptic plasticity, in contrast to the stress effects by micromolar CORT from adrenal glands.

    DOI: 10.1371/journal.pone.0021631

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0003-2044-316X

  • Hippocampal synthesis of sex steroids and corticosteroids: essential for modulation of synaptic plasticity. Reviewed International journal

    Yasushi Hojo, Shimpei Higo, Suguru Kawato, Yusuke Hatanaka, Yuuki Ooishi, Gen Murakami, Hirotaka Ishii, Yoshimasa Komatsuzaki, Mari Ogiue-Ikeda, Hideo Mukai, Tetsuya Kimoto

    Frontiers in endocrinology   2 ( OCT )   43 - 43   2011

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Sex steroids play essential roles in the modulation of synaptic plasticity and neuroprotection in the hippocampus. Accumulating evidence shows that hippocampal neurons synthesize both estrogen and androgen. Recently, we also revealed the hippocampal synthesis of corticosteroids. The accurate concentrations of these hippocampus-synthesized steroids are determined by liquid chromatography-tandem mass-spectrometry in combination with novel derivatization. The hippocampal levels of 17β-estradiol (E2), testosterone (T), dihydrotestosterone (DHT), and corticosterone (CORT), are 5-15 nM, and these levels are sufficient to modulate synaptic plasticity. Hippocampal E2 modulates memory-related synaptic plasticity not only slowly/genomically but also rapidly/non-genomically. Slow actions of E2 occur via classical nuclear receptors (ERα or ERβ), while rapid E2 actions occur via synapse-localized or extranuclear ERα or ERβ. Nanomolar concentrations of E2 change rapidly the density and morphology of spines in hippocampal neurons. ERα, but not ERβ, drives this enhancement/suppression of spinogenesis in adult animals. Nanomolar concentrations of androgens (T and DHT) and CORT also increase the spine density. Kinase networks are involved downstream of ERα and androgen receptor. Newly developed Spiso-3D mathematical analysis is useful to distinguish these complex effects by sex steroids and kinases. Significant advance has been achieved in investigations of rapid modulation by E2 of the long-term depression or the long-term potentiation.

    DOI: 10.3389/fendo.2011.00043

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0003-2044-316X

  • Semicomprehensive analysis of the postnatal age-related changes in the mRNA expression of sex steroidogenic enzymes and sex steroid receptors in the male rat hippocampus. Reviewed International journal

    Tetsuya Kimoto, Hirotaka Ishii, Shimpei Higo, Yasushi Hojo, Suguru Kawato

    Endocrinology   151 ( 12 )   5795 - 806   2010.12

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:ENDOCRINE SOC  

    Although sex steroids play a crucial role in the postnatal brain development, the age-related changes in the hippocampal steroidogenesis remain largely unknown. We performed comprehensive investigations for the mRNA expressions of 26 sex steroidogenic enzymes/proteins and three sex steroid receptors in the male rat hippocampus, at the ages of postnatal day (PD) 1, PD4, PD7, PD10, PD14, 4 wk, and 12 wk (adult), by RT-PCR/Southern blotting analysis. The relative expression levels of these enzymes/receptors at PD1 were Srd5a1 > Star > Ar ∼ Hsd17b4 ∼ Hsd17b1 ∼ Hsd17b7 ∼ Esr1 ∼ Srd5a2 > Hsd17b3 > Esr2 > Cyp11a1 > Cyp17a1 > Cyp19a1 ∼ Hsd17b2 > 3β-hydroxysteroid dehydrogenase I. The mRNA levels of essential enzymes for progesterone/testosterone/estradiol metabolisms (Cyp17a1, Hsd17b7, and Cyp19a1) were approximately constant between PD1 and PD14 and then declined toward the adult levels. Cyp11a1 increased during PD4-PD14 and then considerably decreased toward the adult level (∼8% of PD1). Hsd17b1, Hsd17b2, and 3β-hydroxysteroid dehydrogenase I mRNA decreased approximately monotonously. Hsd17b3 increased to approximately 200% of PD1 during PD4-PD14 and was maintained at this high level. The 5α-reductase mRNA was maintained constant (Srd5a1) or decreased monotonically (Srd5a2) toward the adult level. The Esr1 level peaked at PD4 and decreased toward the adult level, whereas Ar greatly increased during PD1-PD14 and was maintained at this high level. The Star and Hsd17b4 levels were maintained constant from neonate to adult. These results suggest that the hippocampal sex steroidogenic properties are substantially altered during the postnatal development processes, which might contribute to brain sexual maturation.

    DOI: 10.1210/en.2010-0581

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0003-2044-316X

  • Voltage-gated Ca2+ channel mRNAs and T-type Ca2+ currents in rat gonadotropin-releasing hormone neurons. Reviewed

    Nobuyuki Tanaka, Hirotaka Ishii, Chengzhu Yin, Makiko Koyama, Yasuo Sakuma, Masakatsu Kato

    The journal of physiological sciences : JPS   60 ( 3 )   195 - 204   2010.5

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:SPRINGER TOKYO  

    Gonadotropin-releasing hormone (GnRH) neurons play a pivotal role in the neuroendocrine regulation of reproduction. We have previously reported that rat GnRH neurons exhibit voltage-gated Ca(2+) currents. In this study, oligo-cell RT-PCR was carried out to identify subtypes of the alpha(1) subunit of voltage-gated Ca(2+) channels in adult rat GnRH neurons. GnRH neurons expressed mRNAs for all five types of voltage-gated Ca(2+) channels. For T-type Ca(2+) channels, alpha(1H) was dominantly expressed in GnRH neurons. Electrophysiological analysis in acute slice preparations revealed that GnRH neurons from adult rats exhibited T-type Ca(2+) currents with fast inactivation kinetics (~20 ms at -30 mV) and a time constant of recovery from inactivation of ~0.6 s. These results indicate that rat GnRH neurons express subtypes of the alpha(1) subunit for all five types of voltage-gated Ca(2+) channel, and that alpha(1H) was the dominant subtype in T-type Ca(2+) channels.

    DOI: 10.1007/s12576-010-0085-z

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0003-2044-316X

  • Alternative promoter usage and alternative splicing of the rat estrogen receptor alpha gene generate numerous mRNA variants with distinct 5'-ends. Reviewed International journal

    Hirotaka Ishii, Momoko Kobayashi, Yasuo Sakuma

    The Journal of steroid biochemistry and molecular biology   118 ( 1-2 )   59 - 69   2010.1

     More details

    Authorship:Lead author, Corresponding author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:PERGAMON-ELSEVIER SCIENCE LTD  

    The 5'-untranslated region (UTR) of the estrogen receptor alpha (ERalpha) gene plays an important role in determining tissue-specific expression. To elucidate the regulatory mechanisms of rat ERalpha gene expression, the genomic organization must be investigated. We therefore analyzed the structure of the rat ERalpha mRNA 5'-UTR using rapid amplification of 5'-cDNA ends (5'-RACE) and RT-PCR. The analysis showed the presence of multiple variants containing unique 5'-UTRs. We mapped the cDNA sequences on the rat genome, and newly identified one leader exon (exon 0U) and ten untranslated internal exons (exons I1-10). Both splicing from four different leader exons (exons 0S, 0N, 0U, and 0/B) onto exon 1 and alternative splicing in combination with eleven internal exons (exons I1-10, and 0T) produce multiple transcripts. RT-PCR analysis revealed that each variant had preferred expression sites, suggesting that promoter usage and splicing are regulated in tissue-specific manners. Moreover, we determined a splicing event to yield Deltaexon 1 variants (0S-2-3-4-5-6-7-8), which are translated into rat 46 kDa ERalpha proteins. Our results indicate that the rat ERalpha gene is more complex than previously thought in terms of genomic organization and that both alternative promoter usage and alternative splicing contribute to the remarkable diversity of ERalpha mRNAs.

    DOI: 10.1016/j.jsbmb.2009.10.001

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0003-2044-316X

  • Comparison between hippocampus-synthesized and circulation-derived sex steroids in the hippocampus. Reviewed International journal

    Yasushi Hojo, Shimpei Higo, Hirotaka Ishii, Yuuki Ooishi, Hideo Mukai, Gen Murakami, Toshihiro Kominami, Tetsuya Kimoto, Seijiro Honma, Donald Poirier, Suguru Kawato

    Endocrinology   150 ( 11 )   5106 - 12   2009.11

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:ENDOCRINE SOC  

    Estradiol (E2) and other sex steroids play essential roles in the modulation of synaptic plasticity and neuroprotection in the hippocampus. To clarify the mechanisms for these events, it is important to determine the respective role of circulating vs. locally produced sex steroids in the male hippocampus. Liquid chromatography-tandem mass spectrometry in combination with novel derivatization was employed to determine the concentration of sex steroids in adult male rat hippocampus. The hippocampal levels of 17beta-E2, testosterone (T), and dihydrotestosterone (DHT) were 8.4, 16.9, and 6.6 nm, respectively, and these levels were significantly higher than circulating levels. The hippocampal estrone (E1) level was, in contrast, very low around 0.015 nm. After castration to deplete circulating high level T, hippocampal levels of T and DHT decreased considerably to 18 and 3%, respectively, whereas E2 level only slightly decreased to 83%. The strong reduction in hippocampal DHT resulting from castration implies that circulating T may be a main origin of DHT. In combination with results obtained from metabolism analysis of [(3)H]steroids, we suggest that male hippocampal E2 synthesis pathway may be androstenedione --> T --> E2 or dehydroepiandrosterone --> androstenediol --> T --> E2 but not androstenedione --> E1 --> E2.

    DOI: 10.1210/en.2009-0305

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0003-2044-316X

  • Retinoic acid stimulates 17beta-estradiol and testosterone synthesis in rat hippocampal slice cultures. Reviewed International journal

    Eiji Munetsuna, Yasushi Hojo, Minoru Hattori, Hirotaka Ishii, Suguru Kawato, Atsuhiko Ishida, Shiro A J Kominami, Takeshi Yamazaki

    Endocrinology   150 ( 9 )   4260 - 9   2009.9

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:ENDOCRINE SOC  

    The hippocampus is essentially involved in learning and memory processes. Its functions are affected by various neuromodulators, including 17beta-estradiol, testosterone, and retinoid. Brain-synthesized steroid hormones act as autocrine and paracrine modulators. The regulatory mechanism underlying brain steroidogenesis has not been fully elucidated. Synthesis of sex steroids in the gonads is stimulated by retinoic acids. Therefore, we examined the effects of retinoic acids on estradiol and testosterone biosynthesis in the rat hippocampus. We used cultured hippocampal slices from 10- to 12-d-old male rats to investigate de novo steroidogenesis. The infant rat hippocampus possesses mRNAs for steroidogenic enzymes and retinoid receptors. Slices were used after 24 h of preculture to obtain maximal steroidogenic activity because steroidogenesis in cultured slices decreases with time. The mRNA levels for P450(17alpha), P450 aromatase and estrogen receptor-beta in the slices were increased by treatment with 9-cis-retinoic acid but not by all-trans-isomer. The magnitude of stimulation and the shape of the dose-response curve for the mRNA level for P450(17alpha) were similar to those for cellular retinoid binding protein type 2, the transcription of which is activated by retinoid X receptor signaling. 9-cis-Retinoic acid also induced a 1.7-fold increase in the protein content of P450(17alpha) and a 2-fold increase in de novo synthesis of 17beta-estradiol and testosterone. These steroids may be synthesized from a steroid precursor(s), such as pregnenolone or other steroids, or from cholesterol, as so-called neurosteroids. The stimulation of estradiol and testosterone synthesis by 9-cis-retinoic acid might be caused by activation of P450(17alpha) transcription via retinoid X receptor signaling.

    DOI: 10.1210/en.2008-1644

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0003-2044-316X

  • Comparison of sex-steroid synthesis between neonatal and adult rat hippocampus. Reviewed International journal

    Shimpei Higo, Yasushi Hojo, Hirotaka Ishii, Toshihiro Kominami, Kohei Nakajima, Donald Poirier, Tetsuya Kimoto, Suguru Kawato

    Biochemical and biophysical research communications   385 ( 1 )   62 - 6   2009.7

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:ACADEMIC PRESS INC ELSEVIER SCIENCE  

    Sex-steroid synthesis in the hippocampus had been thought to be much more active at the neonatal stage than at the adult stage. However, the detailed comparison between these two stages had not been demonstrated yet. Here we performed the comparison about the mRNA level of steroidogenic enzymes and the rate of steroid metabolism between these two stages of the hippocampus. The relative expression level of P450(17alpha), 17beta- or 3beta-hydroxysteroid dehydrogenase, or P450arom was approximately 1.3-1.5-fold higher at the neonatal than at the adult stage. The rate of sex-steroid metabolism (from dehydroepiandrosterone to estradiol) was 2-7-fold (depending on different steps) more rapid at the neonatal than at the adult stage. Taken together, neonatal steroidogenesis is moderately more active than adult steroidogenesis.

    DOI: 10.1016/j.bbrc.2009.05.005

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0003-2044-316X

  • Ca2+ channels and Ca2+-activated K+ channels in adult rat gonadotrophin-releasing hormone neurones. Reviewed International journal

    M Kato, N Tanaka, H Ishii, C Yin, Y Sakuma

    Journal of neuroendocrinology   21 ( 4 )   312 - 5   2009.3

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:WILEY-BLACKWELL  

    Gonadotrophin-releasing hormone (GnRH) neurones represent the final output neurones in the neuroendocrine system for the control of reproduction. To understand the reproductive neuroendocrine system, an investigation of the intrinsic and extrinsic properties of GnRH neurones is essential. In this review, we focus on the intrinsic properties and summarise our recent findings of ion channels expressed in rat GnRH neurones. Rat GnRH neurones express all four types of high voltage-activated Ca(2+) channel (L, N, P/Q, R) and the low voltage-activated Ca(2+) channel (T). GnRH neurones also express two types of Ca(2+)-activated K(+) [K(Ca)] channel: the small conductance Ca(2+)-activated K(+) (SK) channel and the large conductance Ca(2+)- and voltage-activated K(+) (BK) channel. The activities of these Ca(2+) and K(Ca) channels regulate cell excitability and cellular calcium homeostasis.

    DOI: 10.1111/j.1365-2826.2009.01849.x

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0003-2044-316X

  • Gene structures, biochemical characterization and distribution of rat melatonin receptors. Reviewed

    Hirotaka Ishii, Nobuyuki Tanaka, Momoko Kobayashi, Masakatsu Kato, Yasuo Sakuma

    The journal of physiological sciences : JPS   59 ( 1 )   37 - 47   2009.1

     More details

    Authorship:Lead author, Corresponding author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:SPRINGER TOKYO  

    G-protein coupled receptors for the pineal hormone melatonin have been partially cloned from rats. However, insufficient information about their cDNA sequences has hindered studies of their distribution and physiological responses to melatonin using rats as an animal model. We have cloned cDNAs of two rat membrane melatonin receptor subtypes, melatonin receptor 1a (MT1) and melatonin receptor 1b (MT2), using a rapid amplification of cDNA end (RACE) method. The rat MT1 and MT2 cDNAs encode proteins of 353 and 364 amino acids, respectively, and show 78-93% identities with the human and mouse counterparts. Stable expression of either rat MT1 or MT2 in NIH3T3 cells resulted in high affinity 2-[(125)I]-iodomelatonin ((125)I-Mel) binding (K (d) = 73.2 +/- 9.0 and 73.7 +/- 2.9 pM, respectively), and exhibited a similar rank order of inhibition of specific (125)I-Mel binding by five ligands (2-iodomelatonin > melatonin > 6-hydroxymelatonin > luzindole > N-acetyl-5-hydroxytryptamine). RT-PCR analysis showed that MT1 is highly expressed in the hypothalamus, lung, kidney, adrenal gland, stomach, and ovary, while MT2 is highly expressed in the hippocampus, kidney, and ovary. We also performed multi-cell RT-PCR to examine the expression of mRNAs encoding MT1 and MT2 in adult GnRH neurons. MT1 was weakly expressed in male GnRH neurons, and was less expressed in the female neurons. MT2 expression was undetectable in GnRH neurons from either sex. This study delineates the gene structures, fundamental properties, and distribution of both rat melatonin receptor subtypes, and may offer opportunities to assess the physiological significance of melatonin in rats.

    DOI: 10.1007/s12576-008-0003-9

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0003-2044-316X

  • Cetrorelix, a gonadotropin-releasing hormone antagonist, induces the expression of melatonin receptor 1a in the gonadotropin-releasing hormone neuronal cell line GT1-7. Reviewed International journal

    Hirotaka Ishii, Shun Sato, Chengzhu Yin, Yasuo Sakuma, Masakatsu Kato

    Neuroendocrinology   90 ( 3 )   251 - 9   2009

     More details

    Authorship:Lead author, Corresponding author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:KARGER  

    Melatonin has been implicated in the control of the reproductive system, and the modulatory actions of melatonin on gonadotropin-releasing hormone (GnRH) neurons have been assumed to be indirectly mediated through afferent neurons. However, our previous studies demonstrate sexually dimorphic modulation of A-type gamma-aminobutyric acid (GABA) receptor (GABA(A)R) currents by melatonin in adult rat GnRH neurons and a preferential expression of melatonin 1a receptor (MT1) in male GnRH neurons. Using immortalized GnRH neurons (GT1-7 cells), the present study investigated the mechanism by which the expression of melatonin receptors is regulated in GnRH neurons. Like endogenous GnRH neurons, GT1-7 cells express both GnRH and GnRH receptor mRNAs, indicating that the cells have a self-stimulatory system. A 2-iodomelatonin binding assay and RT-PCR analysis demonstrated that the cells expressed neither MT1 nor MT2. However, treatment of GT1-7 cells with the GnRH antagonist cetrorelix significantly increased 2-iodomelatonin binding and induced a time- and concentration-dependent MT1 mRNA expression. The GABA(A)R currents were then measured using a perforated patch-clamp technique to examine whether the treatment with cetrorelix changed the responses to melatonin. Melatonin augmented the GABA(A)R currents in GT1-7 cells treated with 1 muM cetrorelix for 24 h, while melatonin decreased the currents in the cells not treated with cetrorelix, probably via receptor-independent processes. The present results suggest that GnRH downregulates the expression of MT1 via an autocrine-paracrine mechanism in GT1-7 cells, and modifies the melatonin-induced modulation of GABA(A)R currents. These findings may provide one possible mechanism for the sexually dimorphic responses to melatonin in adult rat GnRH neurons.

    DOI: 10.1159/000231993

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0003-2044-316X

  • Estrogen synthesis in the brain--role in synaptic plasticity and memory. Reviewed International journal

    Yasushi Hojo, Gen Murakami, Hideo Mukai, Shimpei Higo, Yusuke Hatanaka, Mari Ogiue-Ikeda, Hirotaka Ishii, Tetsuya Kimoto, Suguru Kawato

    Molecular and cellular endocrinology   290 ( 1-2 )   31 - 43   2008.8

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:ELSEVIER IRELAND LTD  

    Estrogen and androgen are synthesized from cholesterol locally in hippocampal neurons of adult animals. These neurosteroids are synthesized by cytochrome P450s and hydroxysteroid dehydrogenases (HSDs) and 5alpha-reductase. The expression levels of enzymes are as low as 1/200-1/50,000 of those in endocrine organs, however these numbers are high enough for local synthesis. Localization of P450(17alpha), P450arom, 17beta-HSD and 5alpha-reductase is observed in principal glutamatergic neurons in CA1, CA3 and the dendate gyrus. Several nanomolar levels of estrogen and androgen are observed in the hippocampus. Estrogen modulates memory-related synaptic plasticity not only slowly but also rapidly in the hippocampus. Rapid action of 17beta-estradiol via membrane receptors is demonstrated for spinogenesis and long-term depression (LTD). The enhancement of LTD by 1-10nM estradiol occurs within 1 h. The density of spine is increased in CA1 pyramidal neurons within 2h after application of estradiol. The density of spine-like structure is, however, decreased by estradiol in CA3 pyramidal neurons. ERalpha, but not ERbeta, induces the same enhancement/suppression effects on both spinogenesis and LTD.

    DOI: 10.1016/j.mce.2008.04.017

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0003-2044-316X

  • Activation of A-type gamma-amino butyric acid receptors excites gonadotrophin-releasing hormone neurones isolated from adult rats. Reviewed International journal

    C Yin, H Ishii, N Tanaka, Y Sakuma, M Kato

    Journal of neuroendocrinology   20 ( 5 )   566 - 75   2008.5

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:BLACKWELL PUBLISHING  

    Gonadotrophin-releasing hormone (GnRH) neurones represent the final output neurones in the neuroendocrine control of reproduction, and gamma-amino butyric acid (GABA) is one of the major players in the regulation of GnRH neurones. GABA inhibits a large proportion of brain neurones in adult animals by acting on A-type GABA receptors (GABA(A)Rs). Two contradictory reports on the action of GABA in the GnRH neurones of adult mice have been published. DeFazio et al. (Mol Endocrinol 2002; 16: 2872) demonstrated that activation of GABA(A)Rs excites the GnRH neurones of adult mice, whereas Han et al. (Endocrinology 2002; 143: 1459) showed that the response to GABA on GnRH neurones switches from depolarisation to hyperpolarisation around puberty in female mice. Therefore, we examined the reversal potential of GABA(A)R currents by means of perforated patch-clamp recording with gramicidin in overnight-cultured GnRH neurones isolated from adult GnRH-enhanced green fluorescent protein transgenic rats. The reversal potential was -26 +/- 1.4 mV (mean +/- SEM, n = 42) in GnRH neurones, whereas it was -57 +/- 2.7 mV (n = 34) in unidentified neurones, and GABA depolarised the GnRH neurones in current-clamp condition. The GABA(A)R currents in rat GnRH neurones were augmented by neurosteroids, allopregnanolone and 3 alpha,21-dihydroxy-5 alpha-pregnan-20-one, at submicromolar concentrations. In addition, the expression patterns of GABA(A)R subunit mRNAs were determined by multi-cell reverse transcription-polymerase chain reaction, which revealed that the alpha2, beta 3, gamma 1 and gamma 2 subunits were dominant and the alpha 6 and gamma 3 subunits were negative in rat GnRH neurones. These results indicate that GABA(A)Rs in the soma of rat GnRH neurones are comprised mainly of alpha2, beta 3 and gamma 1 or gamma 2 subunits and that they are sensitive to neurosteroids; moreover, they suggest that activation of these receptors depolarises GnRH neurones. Thus, GABA and neurosteroids influence the electrical activity of GnRH neurones.

    DOI: 10.1111/j.1365-2826.2008.01697.x

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0003-2044-316X

  • 17beta-estradiol at physiological concentrations augments Ca(2+) -activated K+ currents via estrogen receptor beta in the gonadotropin-releasing hormone neuronal cell line GT1-7. Reviewed International journal

    Ichiro Nishimura, Kumiko Ui-Tei, Kaoru Saigo, Hirotaka Ishii, Yasuo Sakuma, Masakatsu Kato

    Endocrinology   149 ( 2 )   774 - 82   2008.2

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:ENDOCRINE SOC  

    Estrogens play essential roles in the neuroendocrine control of reproduction. In the present study, we focused on the effects of 17beta-estradiol (E2) on the K(+) currents that regulate neuronal cell excitability and carried out perforated patch-clamp experiments with the GnRH-secreting neuronal cell line GT1-7. We revealed that a 3-d incubation with E2 at physiological concentrations (100 pm to 1 nm) augmented Ca(2+)-activated K(+) [K(Ca)] currents without influencing Ca(2+)-insensitive voltage-gated K(+) currents in GT1-7 cells. Acute application of E2 (1 nm) had no effect on the either type of K(+) current. The augmentation was completely blocked by an estrogen receptor (ER) antagonist, ICI-182,780. An ERbeta-selective agonist, 2,3-bis(4-hydroxyphenyl)-propionitrile, augmented the K(Ca) currents, although an ERalpha-selective agonist, 4,4',4''-[4-propyl-(1H)-pyrazole-1,3,5-triyl]tris-phenol, had no effect. Knockdown of ERbeta by means of RNA interference blocked the effect of E2 on the K(Ca) currents. Furthermore, semiquantitative RT-PCR analysis revealed that the levels of BK channel subunit mRNAs for alpha and beta4 were significantly increased by incubating cells with 300 pm E2 for 3 d. In conclusion, E2 at physiological concentrations augments K(Ca) currents through ERbeta in the GT1-7 GnRH neuronal cell line and increases the expression of the BK channel subunit mRNAs, alpha and beta4.

    DOI: 10.1210/en.2007-0759

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0003-2044-316X

  • Rat GnRH neurons exhibit large conductance voltage- and Ca2+-Activated K+ (BK) currents and express BK channel mRNAs. Reviewed

    Yoshie Hiraizumi, Ichiro Nishimura, Hirotaka Ishii, Nobuyuki Tanaka, Toshiyuki Takeshita, Yasuo Sakuma, Masakatsu Kato

    The journal of physiological sciences : JPS   58 ( 1 )   21 - 9   2008.2

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:PHYSIOLOGICAL SOC JAPAN  

    Gonadotropin-releasing hormone (GnRH) neurons form the final common pathway for the central regulation of reproduction. As in other neurons, the discharge pattern of action potentials is important for these neurons to function properly. Therefore it is important to elucidate the expression patterns of various types of ion channels in these neurons because they determine cell excitability. To date, voltage-gated Ca2+ channels and SK channels have been reported to be expressed in rat GnRH neurons. In this study, we focused on K+ channels and analyzed their expression in primary cultured GnRH neurons, prepared from GnRH-EGFP transgenic rats, by means of perforated patch-clamp recordings. GnRH neurons exhibited delayed-rectifier K+ currents and large conductance voltage- and Ca2+-activated K+ (BK) currents. Moreover, multicell RT-PCR (reverse transcriptase-polymerase chain reaction) experiments revealed the expression of BK channel mRNAs (alpha, beta1, beta2, and beta4). The results show the presence of delayed-rectifier K+ currents and BK currents besides previously reported slow afterhyperpolarization currents. These currents control the action potential repolarization and probably also the firing pattern, thereby regulating the cell excitability of GnRH neurons.

    DOI: 10.2170/physiolsci.RP013207

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0003-2044-316X

  • Local production of estrogen and its rapid modulatory action on synaptic plasticity Reviewed

    Suguru Kawato, Yasushi Hojo, Hideo Mukai, Gen Murakami, Mari Ogiue-Ikeda, Hirotaka Ishii, Tetsuya Kimoto

    Neuroactive Steroids in Brain Function, Behavior and Neuropsychiatric Disorders: Novel Strategies for Research and Treatment   143 - 169   2008

     More details

    Language:English   Publishing type:Part of collection (book)   Publisher:Springer Netherlands  

    It has long been a common sense that sex hormones are synthesized in the gonads, and reach the brain via the blood circulation. In contrast with this view, the authors demonstrate that estrogen and androgen are also synthesized locally in the hippocampus of adult animals, from cholesterol through dehydroepiandrosterone in hippocampal neurons. These neurosteroids are synthesized by cytochrome P450s and hydroxysteroid dehydrogenases and 5α-reductase. The expression levels of enzymes are as low as 1/200-1/50,000 of those in endocrine organs, preventing quantitative investigations. Localization of P450(17α) and P450arom is observed in synapses of principal glutamatergic neurons, in addition to endoplasmic reticulum, suggesting synaptocrine machanisms. Because several nanomolar estrogen and androgen are observed in the hippocampus, they are expected to have physiological functions. Estrogen modulates memory-related synaptic plasticity not only slowly, but also rapidly in the hippocampus. Molecular mechanisms of rapid action via membrane receptors have not been well elucidated in comparison with those of delayed action via genomic processes. We here describe rapid modulation of representative synaptic plasticity, i.e., long-term depression (LTD), long-term potentiation (LTP) and spinogenesis, by 17β-estradiol, selective estrogen agonists. We demonstrate that 1-10 nM estradiol induced rapid enhancement of LTD within 1 h in CA1, CA3 and dentate gyrus (DG). On the other hand, the modulation of LTP by estradiol is not statistically significant. The total density of spines is increased in CA1 pyramidal neurons, within 2 h after application of estradiol. The total density of thorns (postsynaptic spine-like structure) is, however, decreased by estradiol in CA3 pyramidal neurons. Both the increase of spines in CA1 and the decrease of thorns in CA3 are driven by Erk MAP kinase. Only agonist of estrogen receptor ERalpha induces the same enhancement/suppression effect as estradiol on both LTD and spinogenesis in CA1 and CA3. ERbeta agonist induces completely different results. Estrogen receptor ERalpha localizes in spines and presynapses of principal glutamatergic neurons in CA1, CA3 and DG. The same ERalpha is also located in nuclei. Identification of ERalpha is successfully performed using purified RC-19 antibody. Attention must be paid to the fact that non-purified ERalpha antisera often react significantly with unknown proteins, resulting in wrong staining different from real ERalpha distribution. Identification of kinases/phosphatases in downstream of ERalpha as well as other synaptic estrogen receptors is essential to advance the field. © 2008 Springer Netherlands.

    DOI: 10.1007/978-1-4020-6854-6_7

    Scopus

    researchmap

    Other Link: http://orcid.org/0000-0003-2044-316X

  • Local production of sex hormones and their modulation of hippocampal synaptic plasticity. Reviewed International journal

    Hirotaka Ishii, Tomokazu Tsurugizawa, Mari Ogiue-Ikeda, Makoto Asashima, Hideo Mukai, Gen Murakami, Yasushi Hojo, Tetsuya Kimoto, Suguru Kawato

    The Neuroscientist : a review journal bringing neurobiology, neurology and psychiatry   13 ( 4 )   323 - 34   2007.8

     More details

    Authorship:Lead author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:SAGE PUBLICATIONS INC  

    It is believed that sex hormones are synthesized in the gonads and reach the brain via the blood circulation. In contrast with this view, the authors have demonstrated that sex hormones are also synthesized locally in the hippocampus and that these steroids act rapidly to modulate neuronal synaptic plasticity. The authors demonstrated that estrogens are locally synthesized from cholesterol through dehydroepiandrosterone and testosterone in adult hippocampal neurons. Significant expression of mRNA for P450(17alpha), P450arom, and other steroidogenic enzymes was demonstrated. Localization of P450(17alpha) and P450arom was observed in synapses of principal neurons. In contrast to the slow action of gonadal estradiol, hippocampal neuron-derived estradiol may act locally and rapidly within the neurons. For example, 1 to 10 nM estradiol rapidly enhances long-term depression (LTD). The density of thin spines is selectively increased within two hours upon application of estradiol in pyramidal neurons. Estrogen receptor ERalpha agonist has the same enhancing effect as estradiol on both LTD and spinogenesis. Localization of ERalpha in spines in addition to nuclei of principal neurons implies that synaptic ERalpha is responsible for rapid modulation of synaptic plasticity by endogenous estradiol. Activin A, a peptide sex hormone, may also play a role as a local endogenous modulator of synaptic plasticity.

    DOI: 10.1177/10738584070130040601

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0003-2044-316X

  • Rapid modulation of long-term depression and spinogenesis via synaptic estrogen receptors in hippocampal principal neurons. Reviewed International journal

    Hideo Mukai, Tomokazu Tsurugizawa, Gen Murakami, Shiro Kominami, Hirotaka Ishii, Mari Ogiue-Ikeda, Norio Takata, Nobuaki Tanabe, Aizo Furukawa, Yasushi Hojo, Yuuki Ooishi, John H Morrison, William G M Janssen, John A Rose, Pierre Chambon, Shigeaki Kato, Shunsuke Izumi, Takeshi Yamazaki, Tetsuya Kimoto, Suguru Kawato

    Journal of neurochemistry   100 ( 4 )   950 - 67   2007.2

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:WILEY  

    Rapid modulation of hippocampal synaptic plasticity by estrogen has long been a hot topic, but analysis of molecular mechanisms via synaptic estrogen receptors has been seriously difficult. Here, two types of independent synaptic plasticity, long-term depression (LTD) and spinogenesis, were investigated, in response to 17beta-estradiol and agonists of estrogen receptors using hippocampal slices from adult male rats. Multi-electrode investigations demonstrated that estradiol rapidly enhanced LTD not only in CA1 but also in CA3 and dentate gyrus. Dendritic spine morphology analysis demonstrated that the density of thin type spines was selectively increased in CA1 pyramidal neurons within 2 h after application of 1 nm estradiol. This enhancement of spinogenesis was completely suppressed by mitogen-activated protein (MAP) kinase inhibitor. Only the estrogen receptor (ER) alpha agonist, (propyl-pyrazole-trinyl)tris-phenol (PPT), induced the same enhancing effect as estradiol on both LTD and spinogenesis in the CA1. The ERbeta agonist, (4-hydroxyphenyl)-propionitrile (DPN), suppressed LTD and did not affect spinogenesis. Because the mode of synaptic modulations by estradiol was mostly the same as that by the ERalpha agonist, a search was made for synaptic ERalpha using purified RC-19 antibody qualified using ERalpha knockout (KO) mice. Localization of ERalpha in spines of principal glutamatergic neurons was demonstrated using immunogold electron microscopy and immunohistochemistry. ERalpha was also located in nuclei, cytoplasm and presynapses.

    DOI: 10.1111/j.1471-4159.2006.04264.x

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0003-2044-316X

  • Enhancement of nitric oxide production by association of nitric oxide synthase with N-methyl-D-aspartate receptors via postsynaptic density 95 in genetically engineered Chinese hamster ovary cells: real-time fluorescence imaging using nitric oxide sensitive dye. Reviewed International journal

    Hirotaka Ishii, Keisuke Shibuya, Yoshihiro Ohta, Hideo Mukai, Shigeo Uchino, Norio Takata, John A Rose, Suguru Kawato

    Journal of neurochemistry   96 ( 6 )   1531 - 9   2006.3

     More details

    Authorship:Lead author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:BLACKWELL PUBLISHING  

    The current quantitative study demonstrates that the recruitment of neuronal nitric oxide synthase (nNOS) beneath N-methyl-D-aspartate (NMDA) receptors, via postsynaptic density 95 (PSD-95) proteins significantly enhances nitric oxide (NO) production. Real-time single-cell fluorescence imaging was applied to measure both NO production and Ca(2+) influx in Chinese hamster ovary (CHO) cells expressing recombinant NMDA receptors (NMDA-R), nNOS, and PSD-95. We examined the relationship between the rate of NO production and Ca(2+) influx via NMDA receptors using the NO-reactive fluorescent dye, diaminofluorescein-FM (DAF-FM) and the Ca(2+)-sensitive yellow cameleon 3.1 (YC3.1), conjugated with PSD-95 (PSD-95-YC3.1). The presence of PSD-95 enhanced the rate of NO production by 2.3-fold upon stimulation with 100 microm NMDA in CHO1(+) cells (expressing NMDA-R, nNOS and PSD-95) when compared with CHO1(-) cells (expressing NMDA-R and nNOS lacking PSD-95). The presence of nNOS inhibitor or NMDA-R blocker almost completely suppressed this NMDA-stimulated NO production. The Ca(2+) concentration beneath the NMDA-R, [Ca(2+)](NR), was determined to be 5.4 microm by stimulating CHO2 cells (expressing NMDA-R and PSD-95-YC3.1) with 100 microm NMDA. By completely permealizing CHO1 cells with ionomycin, a general relationship curve of the rate of NO production versus the Ca(2+) concentration around nNOS, [Ca(2+)](NOS), was obtained over the wide range of [Ca(2+)](NOS). This sigmoidal curve had an EC(50) of approximately 1.2 microm of [Ca(2+)](NOS), implying that [Ca(2+)](NR) = 5.4 microm can activate nNOS effectively.

    DOI: 10.1111/j.1471-4159.2006.03656.x

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0003-2044-316X

  • Local neurosteroid production in the hippocampus: influence on synaptic plasticity of memory. Reviewed International journal

    Hideo Mukai, Tomokazu Tsurugizawa, Mari Ogiue-Ikeda, Gen Murakami, Yasushi Hojo, Hirotaka Ishii, Tetsuya Kimoto, Suguru Kawato

    Neuroendocrinology   84 ( 4 )   255 - 63   2006

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:KARGER  

    In neuroendocrinology, it is believed that steroid hormones are synthesized in the gonads and/or adrenal glands, and reach the brain via the blood circulation. In contrast to this view, we are in progress of demonstrating that estrogens and androgens are also synthesized locally by cytochrome P450s in the hippocampus, and that these steroids act rapidly to modulate neuronal synaptic plasticity. We demonstrated that estrogens were locally synthesized in the adult hippocampal neurons. In the pathway of steroidogenesis, cholesterol is converted to pregnenolone (by P450scc), dehydroepiandrosterone [by P450(17alpha)], androstenediol (by 17beta-hydroxysteroid dehydrogenase, 17beta-HSD), testosterone (by 3beta-HSD) and finally to estradiol (by P450arom) and dihydrotestosterone (by 5alpha-reductase). The basal concentration of estradiol in the hippocampus was approximately 1 nM, which was greater than that in blood plasma. Significant expression of mRNA for P450scc, P450(17alpha), P450arom, 17beta-HSD, 3beta-HSD and 5alpha-reductase was demonstrated by RT-PCR. Their mRNA levels in the hippocampus were 1/200-1/5,000 of those in the endocrine organs. Localization of P450(17alpha) and P450arom was observed in synapses in addition to endoplasmic reticulum of principal neurons using immunoelectron microscopy. Different from slow action of gonadal estradiol which reaches the brain via the blood circulation, hippocampal neuron-derived estradiol may act locally and rapidly within the neurons. For example, 1 nM 17beta-estradiol rapidly enhanced the long-term depression (LTD) not only in CA1 but also in CA3 and dentate gyrus. The density of thin spines was selectively increased within 2 h upon application of 1 nM estradiol in CA1 pyramidal neurons. Only ERalpha agonist propyl-pyrazole-trinyl-phenol induced the same enhancing effect as estradiol on both LTD and spinogenesis in the CA1. ERbeta agonist hydroxyphenyl-propionitrile suppressed LTD and did not affect spinogenesis. Localization of estrogen receptor ERalpha in spines in addition to nuclei of principal neurons implies that synaptic ERalpha can drive rapid modulation of synaptic plasticity by endogenous estradiol.

    DOI: 10.1159/000097747

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0003-2044-316X

  • Hippocampal synthesis of estrogens and androgens which are paracrine modulators of synaptic plasticity: Synaptocrinology Reviewed

    H Mukai, N Takata, HT Ishii, N Tanabe, Y Hojo, A Furukawa, T Kimoto, S Kawato

    NEUROSCIENCE   138 ( 3 )   757 - 764   2006

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:PERGAMON-ELSEVIER SCIENCE LTD  

    Hippocampal pyramidal neurons and granule neurons of adult male rats are equipped with a complete machinery for the synthesis of pregnenolone, dehydroepiandrosterone, testosterone, dihydrotestosterone and 17 beta-estradiol. Both estrogens and androgens are synthesized in male hippocampus. These brain steroids are synthesized by cytochrome P450s (P450scc, P45017 alpha and P450arom), hydroxysteroid dehydrogenases and reductases from endogenous cholesterol. The expression levels of enzymes are as low as 1/300-1/1000 of those in endocrine organs. Synthesis is dependent on the acute Ca2+ influx upon neuron-neuron communication via NMDA receptors. Estradiol is particularly important because estradiol rapidly modulates neuronal synaptic transmission such as long-term potentiation via synaptic estrogen receptors. Xenoestrogens may also act via estrogen-driven signaling pathways. (C) 2005 IBRO. Published by Elsevier Ltd. All rights reserved.

    DOI: 10.1016/j.neuroscience.2005.09.010

    Web of Science

    researchmap

    Other Link: http://orcid.org/0000-0003-2044-316X

  • Role of cytochrome P450 in synaptocrinology: Endogenous estrogen synthesis in the brain hippocampus Reviewed

    Gen Murakami, Nobuaki Tanabe, Hiro-taka Ishii, Mari Ogiue-Ikeda, Tomokazu Tsurugizawa, Hideo Mukai, Yasushi Hojo, Norio Takata, Aizo Furukawa, Tetsuya Kimoto, Suguru Kawato

    DRUG METABOLISM REVIEWS   38 ( 3 )   353 - 369   2006

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:TAYLOR & FRANCIS INC  

    In the hippocampus, the center for learning and memory, cytochrome P450s (P450scc, P450(17 alpha), and P450arom) as well as 17 beta-, 3 beta-hydroxysteroid dehydrogenases, and 5 alpha-reductase participate in the synthesis of brain steroids from endogenous cholesterol. These brain steroids include pregnenolone, dehydroepiandrosterone, testosterone, dihydrotestosterone, and 17 beta-estradiol. Both estrogens and androgens are synthesized in the adult male hippocampal neurons. Although the expression levels of steroidogenic enzymes are as low as 1/200 to 1/50,000 of those in testis or ovary, the levels of synthesized steroids are sufficient for the local usage within small neurons (i.e., intracrine system). This intracrine system contrasts with the endocrine system in which high expression levels of steroidogenic enzymes are necessary in endocrine organs in order to supply steroids to many other organs via blood circulation. Endogenous synthesis of sex steroids in the hypothalamus is also discussed.
    Rapid modulation by estrogens and xenoestrogens is discussed concerning synaptic plasticity such as the long-term potentiation, the long-term depression, or spinogenesis. Synaptic expression of P450(17 alpha), P450arom, and estrogen receptors suggests "synaptocrine mechanisms of brain steroids, which are synthesized at synapses and act as synaptic modulators.

    DOI: 10.1080/03602530600724068

    Web of Science

    researchmap

    Other Link: http://orcid.org/0000-0003-2044-316X

  • Adult male rat hippocampus synthesizes estradiol from pregnenolone by cytochromes P45017 alpha and P450 aromatase localized in neurons Reviewed

    Y Hojo, T Hattori, T Enami, A Furukawa, K Suzuki, HT Ishii, H Mukai, JH Morrison, WGM Janssen, S Kominami, N Harada, T Kimoto, S Kawato

    PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA   101 ( 3 )   865 - 870   2004.1

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:NATL ACAD SCIENCES  

    In adult mammalian brain, occurrence of the synthesis of estradiol from endogenous cholesterol has been doubted because of the inability to detect dehydroepiandrosterone synthase, P45017alpha. In adult male rat hippocampal formation, significant localization was demonstrated for both cytochromes P45017alpha and P450 aromatase, in pyramidal neurons in the CA1-CA3 regions, as well as in the granule cells in the dentate gyrus, by means of immunohistochemical staining of slices. Only a weak immunoreaction of these P450s was observed in astrocytes and oligodendrocytes. ImmunoGold electron microscopy revealed that P45017a and P450 aromatase were localized in pre- and postsynaptic compartments as well as in the endoplasmic reticulum in principal neurons. The expression of these cytochromes was further verified by using Western blot analysis and RT-PCR. Stimulation of hippocampal neurons with N-methyl-D-aspartate induced a significant net production of estradiol. Analysis of radioactive metabolites demonstrated the conversion from [H-3]pregnenolone to [H-3]estradiol through dehydroepiandrosterone and testosterone. This activity was abolished by the application of specific inhibitors of cytochrome P450s. Interestingly, estradiol was not significantly converted to other steroid metabolites. Taken together with our previous finding of a P450scc-containing neuronal system for pregnenolone synthesis, these results imply that 17beta-estradiol is synthesized by P45017alpha and P450 aromatase localized in hippocampal neurons from endogenous cholesterol. This synthesis may be regulated by a glutamate-mediated synaptic communication that evokes Ca2+ signals.

    DOI: 10.1073/pnas.2630225100

    Web of Science

    researchmap

    Other Link: http://orcid.org/0000-0003-2044-316X

  • Hippocampal cytochrome P450s synthesize brain neurosteroids which are paracrine neuromodulators of synaptic signal transduction Reviewed

    Keisuke Shibuya, Norio Takata, Yasushi Hojo, Aizo Furukawa, Nobuaki Yasumatsu, Tetsuya Kimoto, Taihei Enami, Kumiko Suzuki, Nobuaki Tanabe, Hirotaka Ishii, Hideo Mukai, Taiki Takahashi, Taka-aki Hattori, Suguru Kawato

    Biochimica et Biophysica Acta - General Subjects   1619 ( 3 )   301 - 316   2003.2

     More details

    Language:English   Publishing type:Research paper (international conference proceedings)  

    Hippocampal pyramidal neurons and granule neurons of adult male rats are equipped with a complete machinery for the synthesis of pregnenolone, dehydroepiandrosterone, 17β-estradiol and testosterone as well as their sulfate esters. These brain neurosteroids are synthesized by cytochrome P450s (P450scc, P45017α and P450arom) from endogenous cholesterol. Synthesis is acutely dependent on the Ca2+ influx attendant upon neuron-neuron communication via N-methyl-D-aspartate (NMDA) receptors. Pregnenolone sulfate, estradiol and corticosterone rapidly modulate neuronal signal transduction and the induction of long-term potentiation via NMDA receptors and putative membrane steroid receptors. Brain neurosteroids are therefore promising neuromodulators that may either activate or inactivate neuron-neuron communication, thereby mediating learning and memory in the hippocampus. © 2002 Elsevier Science B.V. All rights reserved.

    DOI: 10.1016/S0304-4165(02)00489-0

    Scopus

    PubMed

    researchmap

▼display all

Books

  • CLINICAL NEUROSCIENCE

    Hirotaka Ishii, Masahiro Morishita( Role: Contributor)

    CHUGAI-IGAKUSHA  2023.8 

     More details

    Language:Japanese   Book type:Scholarly book

    researchmap

  • Histochemistry and Cytochemistry 2023

    Hirotaka Ishii, Morishita Masahiro, Higo Shimpei( Role: ContributorPrinciples and Basics of Immunohistochemistry)

    Japan Society of Histochemistry and Cytochemistry  2023.7 

     More details

  • Neuroactive Steroids in Brain Function, Behavior and Neuropsychiatric Disorders

    Suguru Kawato, Yasushi Hojo, Hideo Mukai, Gen Murakami, Mari Ogiue-Ikeda, Hirotaka Ishii, Tetsuya Kimoto( Role: ContributorLocal Production of Estrogen and its Rapid Modulatory Action on Synaptic Plasticity)

    Springer Dordrecht  2008.2 

     More details

Misc.

▼display all

Presentations

  • C末端欠損型エストロゲン受容体α変異体の恒常的活性化機構の解明

    石井 寛高, 服部 裕次郎, 渡部 寛, 小澤 一史

    日本神経内分泌学会(第42回)  2015.9 

     More details

    Language:Japanese  

    Venue:仙台  

    researchmap

  • ラットエストロゲン受容体α遺伝子のΔexon 1変異体の機能解析

    服部 裕次郎, 石井 寛高, 森田 明夫, 小澤 一史

    第42回日本神経内分泌学会・第23回日本行動神経内分泌研究会・合同学術集会  2015.9 

     More details

    Language:Japanese  

    researchmap

  • Identification of transcriptional and posttranscriptional regulation of human estrogen receptor expression in the testis

    石井 寛高, 服部 裕次郎, 小澤 一史

    日本解剖学会総会・全国学術集会(第120回)  2015.3 

     More details

    Language:English  

    Venue:神戸  

    researchmap

  • 新規ヒトエストロゲン受容体α変異体の同定と転写活性化機構の解明

    服部 裕次郎, 石井 寛高, 森田 明夫, 小澤 一史

    第88回日本内分泌学会学術総会  2015.4 

     More details

    Language:Japanese  

    researchmap

  • 株化細胞を用いた in vitro 吸入麻酔薬作用解析実験系の確立

    永本 盛嗣, 飯島 典生, 相川 優子, 石井 寛高, 肥後 心平, 託見 健, 安齋 めぐみ, 坂本 篤裕, 小澤 一史

    日本神経内分泌学会(第42回)  2015.9 

     More details

    Language:Japanese  

    Venue:仙台  

    researchmap

  • 変形性膝関節症の滑膜ステロイド受容体発現量はBMIと逆相関する

    渡部 寛, 高橋 謙治, 石井 寛高, 小澤 一史, 高井 信朗

    日本軟骨代謝学会(第29回)  2016.2 

     More details

    Language:Japanese  

    Venue:広島  

    researchmap

  • C末端欠損型エストロゲン受容体α変異体の恒常的活性化機構の解明

    石井 寛高, 服部 裕次郎, 渡部 寛, 小澤 一史

    日本神経内分泌学会(第42回)  2015.9 

     More details

    Language:Japanese  

    Venue:仙台  

    researchmap

  • ラットエストロゲン受容体α遺伝子の△exon 1 変異体の機能解析

    服部 裕次郎, 石井 寛高, 森田 明夫, 小澤 一史

    日本神経内分泌学会(第42回)  2015.9 

     More details

    Language:Japanese  

    Venue:仙台  

    researchmap

  • リポソームの細胞内に関する形態学的解析 培養細胞内での局在解析から

    友利 裕二, 日沼 州司, 石井 寛高, 託見 健, 高井 信朗, 小澤 一史

    日本組織細胞化学会総会・学術集会(第55回)  2014.9 

     More details

    Language:Japanese  

    Venue:長野  

    researchmap

  • ヒトエストロゲン受容体βの多重プロモーター機構と 5´-非翻訳領域の選択的スプライシングパターンの同定

    石井 寛高, 服部 裕次郎, 小澤 一史

    日本神経内分泌学会学術集会(第41回)  2014.10 

     More details

    Language:Japanese  

    Venue:東京  

    researchmap

  • 新規ヒトエストロゲン受容体β変異体の同定と機能解析

    服部 裕次郎, 石井 寛高, 森田 明夫, 小澤 一史

    日本神経内分泌学会学術集会 (第41回)  2014.10 

     More details

    Language:Japanese  

    Venue:東京  

    researchmap

  • リポソームの細胞内に関する形態学的解析ー培養細胞内での局在解析からー

    友利 裕二, 飯島 典生, 日沼 州司, 石井 寛高, 託見 健, 高井 信朗, 小澤 一史

    日本組織細胞化学会総会・学術集会 (第55回)  2014.9 

     More details

    Language:Japanese  

    Venue:松本  

    researchmap

  • 新規ヒトエストロゲン受容体β変異体の同定と機能解析

    服部 裕次郎, 石井 寛高, 森田 明夫, 小澤 一史

    日本神経内分泌学会学術集会(第41回)  2014.10 

     More details

    Language:Japanese  

    Venue:東京  

    researchmap

  • 新規ヒトエストロゲン受容体変異体の同定と機能解析

    服部 裕次郎, 石井 寛高, 森田 明夫, 小澤 一史

    臨床内分泌代謝Update(第24回)  2014.11 

     More details

    Language:Japanese  

    Venue:大宮  

    researchmap

  • 麻酔薬の作用機序の研究のための株化細胞を用いたin vitroでの実験系の確立

    永本 盛嗣, 飯島 典夫, 石井 寛高, 託見 健, 坂本 篤裕, 小澤 一史

    日本解剖学会総会・全国学術集会(第120回)  2015.3 

     More details

    Language:Japanese  

    Venue:神戸  

    researchmap

  • ヒトエストロゲン受容体βの多重プロモーター機構と5‘―非翻訳領域の選択的スプライシングパターンの同定

    石井 寛高, 服部 裕次郎, 小澤 一史

    日本神経内分泌学会学術集会 (第41回)  2014.10 

     More details

    Language:Japanese  

    Venue:東京  

    researchmap

  • 新規ヒトエストロゲン受容体変異体の同定と機能解析

    服部 裕次郎, 石井 寛高, 森田 明夫, 小澤 一史

    臨床内分泌代謝Update(第24回)  2014.11 

     More details

    Language:Japanese  

    Venue:大宮  

    researchmap

  • 新規ヒトエストロゲン受容体α変異体の同定と転写活性化機構の解明

    服部 裕次郎, 石井 寛高, 森田 明夫, 小澤 一史

    日本解剖学会総会・全国学術集会(120回)・日本生理学会大会(第92回)合同大会  2015.3 

     More details

    Language:Japanese  

    Venue:神戸  

    researchmap

  • Identification of novel estrogen receptor α variants in the human and mechanism of transcriptional activation

    服部 裕次郎, 石井 寛高, 森田 明夫, 小澤 一史

    日本解剖学会総会・全国学術集会(第120回)  2015.3 

     More details

    Language:English  

    Venue:神戸  

    researchmap

  • GT1-7細胞におけるアセチルコリン受容体の発現解析

    荒井 勇樹, 石井 寬高, 小林 牧人, 小澤 一史

    日本解剖学会総会(第119回)  2014.3 

     More details

    Language:Japanese  

    Venue:栃木  

    researchmap

  • 培養細胞によるリポソーム取り込みメカニズムノ解析

    友利 祐二, 飯島 典生, 日沼 州司, 平井 政彦, 石井 寬高, 託見 健, 高井 信朗, 小澤 一史

    日本解剖学会総会(第119回)  2014.3 

     More details

    Language:Japanese  

    Venue:栃木  

    researchmap

  • 新規ラットC末端欠損型エストロゲン受容体α変異体の同定と機能解析

    服部 裕次郎, 石井 寛高, 森田 明夫, 小澤 一史

    日本神経内分泌学会学術集会(第40回)  2013.10 

     More details

    Language:Japanese  

    Venue:宮崎  

    researchmap

  • 新規ラットC末端欠損型エストリゲン受容体α変異体の同定

    服部 裕次郎, 石井 寬高, 森田 明夫, 小澤 一史

    日本解剖学会総会(第119回)  2014.3 

     More details

    Language:Japanese  

    Venue:栃木  

    researchmap

  • 新規ラットC末端欠損型エストロゲン受容体α変異体の局在・機能解析

    服部 裕次郎, 石井 寛高, 森田 明夫, 小澤 一史

    日本内分泌学会学術総会(第87回)  2014.4 

     More details

    Language:Japanese  

    Venue:福岡  

    researchmap

  • Characterinzation of alternative promoter usage and alternative splicing profiles of the estrogen receptor β gene in the human International conference

    Isii Hirota, Hattori Yuj, Ozawa Hitos

    The International Congress of Neuroendocrinology 2014  2014.8 

     More details

    Language:English  

    Venue:Sydney  

    researchmap

  • 新規ラットC末端欠損型エストロゲン受容体α変異体の同定

    服部 裕次郎, 石井 寛高, 森田 明夫, 小澤 一史

    第119回日本解剖学会総会・全国学術集会  2014.3 

     More details

    Language:Japanese  

    researchmap

  • ラットN末端欠損型エストロゲン受容体の局在・機能解析

    石井 寛高, 服部 裕次郎, 小澤 一史

    日本解剖学会総会(第119回)  2014.3 

     More details

    Language:Japanese  

    Venue:栃木  

    researchmap

  • Identification of novel estrogen receptor β variants in the human International conference

    Hattori Y, Ishii H, Morita A, Ozawa H

    International Congress of Neuroendocrinology (The 8th)  2014.8 

     More details

    Language:English  

    Venue:Sydney, Australia  

    researchmap

  • Identificalion of novel estrogen receptor β variants in the human International conference

    Hattori Yuj, Ishii Hirot, Morita Akio, Ozawa Hitos

    The International Congress of Neuroendocrinology 2014  2014.8 

     More details

    Language:English  

    Venue:Sydney  

    researchmap

  • Identification of truncated estrogen receptor α variants in the mouse International conference

    Kawahigashi, Ishii H, Munetomo A, Hamada T, Sakuma Y

    US-JAPAN Brain Research Cooperative Program Workshop on Prosocial Behavior at Emory University  2011.10 

     More details

    Language:English  

    Venue:Atlanta, GA, U.S.A.  

    researchmap

  • ヒトC末端欠損型エストロゲン受容体αの同定と機能解析

    石井 寛高, 濱田 知宏, 佐久間 康夫

    第38回日本神経内分泌学会学術集会  2011.11 

     More details

    Language:Japanese  

    Venue:東京都  

    researchmap

  • Morphological relationship between gonadotropin-releasing hormone neurons and somatostatin neurons in the organum vasculosum of the lamina terminalis and its physiological meaning

    Koyama M, Yin C, Ishii H, Sakuma Y, Kato M

    The 34th Annual Meeting of the Japan Neuroscience Society  2011.9 

     More details

    Language:English  

    Venue:Yokohama  

    researchmap

  • Slow afterhyperpolarization(sAHP)current in rat GnRH neurons is carried through SK and KCNQ channels

    Yin C, Ishii H, Sakuma Y, Kato M

    The 89th Annual Meeting of the Physiological Society of Japan  2012.3 

     More details

    Language:English  

    Venue:Nagano, Matsumoto  

    researchmap

  • ラットGnRHニューロン及びGT1-7細胞株に発現するGABA受容体の発現解析

    石井 寛高

    日本神経内分泌学会学術集会(第39回)  2012.9 

     More details

    Language:Japanese  

    Venue:北九州  

    researchmap

  • Complex organization of the estrogen receptor αgenes: identification of numerous estrogen receptor αvariants in the human, mouse and rat International conference

    Ishii H, Kobayashi M, Munetomo A, Kajio M, Hamada T, Sakuma Y

    The 7th AOSCE Congress Satellite Symposium Neuroendocrinology and Behaviour  2012.3 

     More details

    Language:English  

    Venue:Penang, Malaysia  

    researchmap

  • Identification of C-terminally-truncated estrogen receptor α variants in the human, mouse and rat

    Ishii H, Munetomo A, Hamada T, Sakuma Y

    The 89th Annual Meeting of the Physiological Society of Japan  2012.3 

     More details

    Language:English  

    Venue:Nagano, Matsumoto  

    researchmap

  • GT1-7細胞におけるアセチルコリン受容体の発現解析

    荒井 勇樹, 石井 寛高, 小林 牧人, 小澤 一史

    日本神経内分泌学会学術集会(第40回)  2013.10 

     More details

    Language:Japanese  

    Venue:宮崎  

    researchmap

  • 新規ラットC末端欠損型エストロゲン受容体α変異体の同定と機能解析

    服部 裕次郎, 石井 寛高, 森田 明夫, 小澤 一史

    第40回日本神経内分泌学会学術集会  2013.10 

     More details

    Language:Japanese  

    researchmap

  • 精巣特異的ヒトエストロゲン受容体α変異体の5’-非翻訳領域は翻訳効率を著しく低減する

    石井 寛高, 小澤 一史

    日本神経内分泌学会学術集会(第40回)  2013.10 

     More details

    Language:Japanese  

    Venue:宮崎  

    researchmap

  • Alternative promoter usage and alternative splicing of the mouse estrogen receptor αgene generate numerous mRNA variants International conference

    Shoda Y, Ishii H, Kobayashi M, Yomogida K, Hamada T, Sakuma Y

    The 7th International Congress Nuroendocrinology  2010.7 

     More details

    Language:English  

    Venue:Rouen, France  

    researchmap

  • GABAA receptors in rat GnRH neurons are composed of α2, β3, γ1-2, and ε subunits International conference

    Yin C, Ishii H, Kato M, Sakuma Y

    The 7th International Congress of Nuroendocrinology  2010.7 

     More details

    Language:English  

    Venue:Rouen, France  

    researchmap

  • イントロン領域で同定された新規エクソンにより,N末端・C末端欠損型エストロゲン受容体α変異体が形成される International conference

    石井 寛高, 庄田 有里, 濱田 知宏, 佐久間 康夫

    第37回日本神経内分泌学会学術集会  2010.10 

     More details

    Language:Japanese  

    Venue:京都  

    researchmap

  • Identification of N-terninally-and C-terminally-truncated estrogen receptor α variants in the human and rat

    Ishii H, Hamada T, Sakuma Y

    第88回日本生理学会・第116回日本解剖学会総会・全国学術集会・合同大会  2011.3 

     More details

    Language:English  

    Venue:横浜  

    researchmap

  • マウスエストロゲン受容体αの新規N末端・C末端欠損型変異体の同定 International conference

    石井 寛高, 庄田 有里, 濱田 知宏, 佐久間 康夫

    愛知県田原市  2010.8 

     More details

    Language:English  

    Venue:愛知県田原市  

    researchmap

  • ラットGnRHニューロンに発現するA型GABA受容体(GABAAR)のサブユニット構成 International conference

    尹 成珠, 石井 寛高, 加藤 昌克, 佐久間 康夫

    第37回日本神経内分泌学会学術集会  2010.10 

     More details

    Language:Japanese  

    Venue:京都  

    researchmap

  • マウスにおける新規C末端欠損型エストロゲン受容体αの同定と機能解析

    河東 堤子, 石井 寛高, 棟朝 亜理紗, 濱田 知宏, 佐久間 康夫

    日本下垂体研究会第26回学術集会  2011.8 

     More details

    Language:Japanese  

    Venue:岡山市  

    researchmap

  • ラットN末端欠損型・C末端欠損型エストロゲン受容体αの同定と機能解析

    石井 寛高, 濱田 知宏, 佐久間 康夫

    日本下垂体研究会第26回学術集会  2011.8 

     More details

    Language:Japanese  

    Venue:岡山市  

    researchmap

  • Distribution of GnRH and somatostatin neuronal fibers in the organum vasculosum of the lamina terminalis and their physiological meaning

    Koyama M, Yin C, Ishii H, Sakuma Y, Kato M

    第88回日本生理学会・第116回日本解剖学会総会・全国学術集会・合同大会  2011.3 

     More details

    Language:English  

    Venue:横浜  

    researchmap

  • T-type calcium channels in rat gonadotropin-releasing hormones

    Tanaka N, Ishii H, Yin C, Koyama M, Sakuma Y

    第88回日本生理学会・第116回日本解剖学会総会・全国学術集会・合同大会  2011.3 

     More details

    Language:English  

    Venue:横浜  

    researchmap

  • Cell-specific usage of an estrogen receptorα gane promoter in the dimorphic nucleus of the rat preoptic aea International conference

    Hamada T, Ishii H, Orikasa C, Sakuma Y

    The Society for Behavioral Neuroendocrinology 14th Annual Meeting  2010.7 

     More details

    Language:English  

    Venue:Toronto, Canada  

    researchmap

  • ラットメラトニン受容体の生化学的解析

    小林 桃子, 石井 寛高, 田中 伸幸, 加藤 昌克, 佐久間 康夫

    第35回日本神経内分泌学会第23回日本下垂体研究会合同学術集会  2008.8 

     More details

    Language:Japanese  

    researchmap

  • γ-Aminobutyric acid depolarizes gonadotropin-releasing hormone (GnRH)neurons in the pre-optic area of adult rats International conference

    Yin C, Tanaka N, Ishii H, Kato M, Sakuma Y

    日本学術振興会二国間交流事業中国とのセミナー学術集会「都市化に代表される社会的変容が脳の健全な育成におよぼす影響についての二国間研究」  2008.10 

     More details

    Language:English  

    Venue:つくば市  

    researchmap

  • Neurosteroids augment A type γ-aminobutyric acid receptor(GABAA) currents in rat GnRH neurons International conference

    Yin C, Ishii H, Tanaka N, Sakuma Y, Kato M

    Congress of the International Union of Physiological Sciences(36th)  2009.7 

     More details

    Language:English  

    Venue:京都  

    researchmap

  • GnRHアンタゴニストのcetrorelixは,GT1-7細胞のメラトニン受容体1aの発現を誘導する

    石井 寛高, 尹 成珠, 加藤 昌克, 佐久間 康夫

    第35回日本神経内分泌学会第23回日本下垂体研究会合同学術集会  2008.8 

     More details

    Language:Japanese  

    researchmap

  • ラットGnRHニューロンにおける電位依存性カルシウムチャネルの発現の発達段階による差位

    田中 伸幸, 石井 寛高, 尹 成珠, 加藤 昌克, 佐久間 康夫

    第35回日本神経内分泌学会第23回日本下垂体研究会合同学術集会  2008.8 

     More details

    Language:Japanese  

    researchmap

  • Neurosterroids augment A-type γ-aminobutyric acid receptor currents in rat gonadotropin-releasing hormone neurons

    Yin C, Ishii H, Tanaka N, Kato M, Sakuma Y

    日本生理学会大会(第87回)  2010.5 

     More details

    Language:Japanese  

    Venue:岩手県盛岡市  

    researchmap

  • Alternative promoter usage and alternative splicing of the estrogen receptor α genes generate numerous mRNA variants

    Ishii H, Kobayashi M, Sakuma Y

    日本生理学会大会(第87回)  2010.5 

     More details

    Language:Japanese  

    Venue:岩手県盛岡市  

    researchmap

  • T-type Ca2+ channels in adult rat gonadotropin-releasing hormone(GnRH) neurons International conference

    Nobuyuki T, Ishii H, Yin C, Sakuma Y, Kato M

    Congress of the International Union of Physiological Sciences(36th)  2009.7 

     More details

    Language:English  

    Venue:京都  

    researchmap

  • Genomic organization and structure of the 5'-flancking region of the rat estrogen receptor alpha gane International conference

    Ishii H, Kobayashi M, Sakuma Y

    Congress of the International Union of Physiological Sciences(36th)  2009.7 

     More details

    Language:English  

    Venue:京都  

    researchmap

  • ラットGnRHニューロンに発現する電位依存性カルシウムチャネルmRNAExpression of voltage-gated Ca <SUP></SUP> channel mRNAs in GnRH neurons of male and female adult rats

    田中 伸幸, 石井 寛高, 尹 成珠, 加藤 昌克, 佐久間 康夫

    第31回日本神経科学学会  2008.7 

     More details

    Language:Japanese  

    researchmap

  • 変形性膝関節症滑膜での最適参照遺伝子の選定に基づいたステロイドホルモン受容体の発現解析と そのBMI,性との相関について

    渡部 寛, 石井 寛高, 高橋 謙治, 高井 信朗, 小澤 一史

    日本解剖学会総会・全国学術集会(第122回)  2017.3 

     More details

    Language:Japanese  

    Venue:長崎  

    researchmap

  • 日本医科大学における実習手順:全体のデザイン,モチベーション,評価

    飯島 典生, 託見 健, 石井 寬高, 岩田 衣世, 楊 春英, 肥後 心平, 小澤 一史

    日本解剖学会総会・全国学術集会(第122回)  2017.3 

     More details

    Language:Japanese  

    Venue:長崎  

    researchmap

  • C末端欠損型性ステロイド受容体変異体の恒常的転写活性化能獲得機構の解明.

    石井寛高, 服部裕次郎, 渡部寛, 小澤一史

    日本解剖学会総会・全国学術集会(第122回)  2017.3 

     More details

    Language:Japanese  

    Venue:長崎  

    researchmap

  • 変形性膝関節症の滑膜ステロイド受容体発現量はBMIと逆相関する

    渡部 寛, 石井 寬高, 高橋 謙治, 高井 信朗, 小澤 一史

    日本解剖学会総会・全国学術集会(第121回)  2016.3 

     More details

    Language:Japanese  

    Venue:郡山  

    researchmap

  • C末端欠損型エストロゲン受容体α変異体の恒常的活性化機構の解析

    服部 裕次郎, 石井 寬高, 渡部 寛, 森田 明夫, 小澤 一史

    日本解剖学会総会・全国学術集会(第121回)  2016.3 

     More details

    Language:Japanese  

    Venue:郡山  

    researchmap

  • 表面電荷の違いによるリポソームの細胞内動態の解析

    友利 裕二, 飯島 典生, 日沼 州司, 天野 千絵, 石井 寬高, 託見 健, 高井 信朗, 小澤 一史

    日本解剖学会総会・全国学術集会(第121回)  2016.3 

     More details

    Language:Japanese  

    Venue:郡山  

    researchmap

  • C末端欠損型エストロゲン受容体α変異体の恒常的活性化にはリガンド結合領域内のヘリックス5の欠損が必須である

    石井 寛高, 服部 裕次郎, 渡辺 寛, 小澤 一史

    日本神経内分泌学会(第43回)・日本行動神経内分泌研究会・合同学術集会(第23回)  2016.10 

     More details

    Language:Japanese  

    researchmap

  • C末端欠損型エストロゲン受容体α変異体の恒常的活性化にはリガンド結合領域内のヘリックス5の欠損が必須である

    石井 寛高, 服部 裕次郎, 渡部 寛, 小澤 一史

    日本神経内分泌学会学術集会(第43回)  2016.10 

     More details

    Language:Japanese  

    Venue:浜松  

    researchmap

  • Steroid Receptors In The Synovial Membranes Of The Human Knee Osteoarthritis:Expression Profiles And Correlations International conference

    Watanabe H, Takahashi K, Ishii H, Iizawa N, Kawaji H, Majima T, Ozawa H, Takai S

    ORS 2016 Annual Meeting  2016.3 

     More details

    Language:English  

    Venue:Orlando  

    researchmap

  • C末端欠損型エストロゲン受容体α変異体の恒常的活性化機構の解析

    服部 裕次郎, 石井 寛高, 渡部 寛, 森田 明夫, 小澤 一史

    日本解剖学会 総会・全国学術集会(第121回)  2016.3 

     More details

    Language:Japanese  

    Venue:福島  

    researchmap

  • C末端欠損型性ステロイド受容体変異体の恒常的転写活性化能獲得機構の解明

    石井 寬高, 服部 裕次郎, 渡部 寛, 小澤 一史

    日本解剖学会総会・全国学術集会(第122回)  2017.3 

     More details

    Language:Japanese  

    Venue:長崎  

    researchmap

  • 変形性膝関節症滑膜での最適参照遺伝子の選定に基づいたステロイドホルモン受容体の発現解析と そのBMI,性との相関について

    渡部 寛, 石井 寛高, 高橋 謙治, 高井 信朗, 小澤 一史

    運動器坑加齢医学研究会(第6回)  2016.11 

     More details

    Language:Japanese  

    Venue:東京  

    researchmap

  • 変形性膝関節症滑膜での最適参照遺伝子の選定に基づいたステロイド受容体の発現解析とそのBMI,性との相関について

    渡部 寛, 石井 寬高, 高橋 謙治, 高井 信朗, 小澤 一史

    日本解剖学会総会・全国学術集会(第122回)  2017.3 

     More details

    Language:Japanese  

    Venue:長崎  

    researchmap

  • Establishment of an in vitro experimental system using a cell line to investigate the mechanisms of anesthesia

    永本 盛嗣, 飯島 典生, 石井 寛高, 託見 健, 坂本 篤裕, 小澤 一史

    日本解剖学会総会・全国学術集会(第120回)  2015.3 

     More details

    Language:English  

    Venue:神戸  

    researchmap

▼display all

Awards

  • 25th Award for Excellent Papers in Acta Histochemica et Cytochemica

    2023.10   Japan Society of Histochemistry and Cytochemistry   Optimized Mouse-on-mouse Immunohistochemical Detection of Mouse ESR2 Proteins with PPZ0506 Monoclonal Antibody

    Mina Ozawa, Yujiro Hattori, Shimpei Higo, Mai Otsuka, Keisuke Matsumoto, Hitoshi Ozawa, Hirotaka Ishii

     More details

  • Kawakami Award

    2021.10   Japan Neuroendocrine Society  

    Hirotaka Ishii

     More details

  • 第24回臨床内分泌代謝Update 優秀ポスター賞

    2014.11   日本内分泌学会   新規ヒトエストロゲン受容体変異体の同定と機能解析

    服部 裕次郎, 石井 寛高, 森田 明夫, 小澤 一史

     More details

  • Hiroshi and Aya Irisawa Memorial Award for Excellent Papers in The Journal of Physiological Sciences

    2011.3   日本生理学会   Voltage-gated Ca2+ channels mRNAs and T-type Ca2+ currents in rat gonadotropin-releasing hormone neurons

    Nobuyuki Tanaka, Hirotaka Ishii, Chengzhu Yin, Makiko Koyama, Yasuo Sakuma, Masakatsu Kato

     More details

  • Hiroshi and Aya Irisawa Memorial Award for Excellent Papers in The Journal of Physiological Sciences

    2010.3   日本生理学会   Gene structures, biochemical characterization and distribution of rat melatonin receptors.

    Hirotaka Ishii, Nobuyuki Tanaka, Momoko Kobayashi, Masakatsu Kato, Yasuo Sakuma

     More details

Research Projects

  • ESR1遺伝子のゲノム構造再編成によるエストロゲン感受性腫瘍増悪機構の解明

    Grant number:23K06350  2023.4 - 2026.3

    日本学術振興会  科学研究費助成事業 基盤研究(C)  基盤研究(C)

    石井 寛高

      More details

    Grant amount:\4550000 ( Direct Cost: \3500000 、 Indirect Cost:\1050000 )

    researchmap

  • Mechanisms of endocrine- and chemotherapy-resistance acquisition in estrogen-sensitive tumors by ESR1 isoforms

    Grant number:18K06879  2018.4 - 2022.3

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research Grant-in-Aid for Scientific Research (C)  Grant-in-Aid for Scientific Research (C)

    Ishii Hirotaka

      More details

    Grant amount:\4160000 ( Direct Cost: \3200000 、 Indirect Cost:\960000 )

    C-terminally truncated estrogen receptor α (ESR1) isoforms have been recognized as dominant negative isoforms. However, our study demonstrated that most of C-terminally truncated ESR1 isoforms retained constitutive transcriptional activity and estrogen receptor (ER) antagonist-insensitivity and that stable transfection of the isoforms into breast cancer-derived cells induced estrogen-independent growth and ER antagonist-resistance. In addition, we identified structure-based mechanisms for the gains-of-function of the ESR1 isoforms.

    researchmap

  • Molecular and morphofunctional study on the novel regulation system for reproduction based on the input of various enviromental information

    Grant number:18K06860  2018.4 - 2021.3

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research Grant-in-Aid for Scientific Research (C)  Grant-in-Aid for Scientific Research (C)

    Ozawa Hitoshi

      More details

    Grant amount:\4420000 ( Direct Cost: \3400000 、 Indirect Cost:\1020000 )

    We have been studying the function and morphology of kisspeptin, a new "main player" in the reproductive regulatory system, and its containing neurons, using histochemical and molecular biological techniques. The results include the relationship between energy metabolism and the reproductive regulatory axis, subtypes of sex hormones and their functions, distribution of kisspeptin-related peptides in the brain and their functions, effects of the maternal-fetal environment on postnatal development based on the Dohad theory, changes in reproductive regulatory mechanisms in animal models of diabetes mellitus, and the effects of kisspeptin on kisspeptin neurons. We have achieved a wide range of research accomplishments.

    researchmap

  • 新規生殖制御系への環境情報入力と3次元的相互作用,生活環境に関する機能形態学的研究

    2016

    科学研究費補助金 基盤研究(C) 

    小澤 一史, 飯島 典生, 託見 健, 石井 寬高, 岩田 衣世, 肥後 心平

      More details

    Grant type:Competitive

    Grant amount:\1200000

    researchmap

  • 新規生殖制御系への環境情報入力と3次元的相互作用,生活環境に関する機能形態学的研究

    2015

    科学研究費補助金 基盤研究(C) 

    小澤 一史, 飯島 典生, 託見 健, 肥後 心平, 石井 寬高, 岩田 衣世

      More details

    Grant type:Competitive

    Grant amount:\1200000

    researchmap

  • 多重プロモーターと選択的スプライシングによるエストロゲン受容体発現調節機構の解明

    2015

    科学研究費補助金 基盤研究(C) 

    石井 寬高

      More details

    Grant type:Competitive

    Grant amount:\1200000

    researchmap

  • Morphofunctional study on inputs of the environmental information, three dimensional interacition and life cycle, of a new system for reproductive regulation.

    Grant number:26460323  2014.4 - 2017.3

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research Grant-in-Aid for Scientific Research (C)  Grant-in-Aid for Scientific Research (C)

    Hitoshi Ozawa

      More details

    Grant amount:\5070000 ( Direct Cost: \3900000 、 Indirect Cost:\1170000 )

    We investigated about the kisspeptin neurons, which is known as the center of reproductive regulation and integrate the several information in the brain to relay to the HPG axis. Actually, we clearly investigated 1) the subcellular localization of kisspepin, NKB, and dynorphin in the KNDy neuronos, which are coexpressed kisspepin, NKB, and dynorphin in the arcute nucleus by immunoelectron microscopy, 2) the interrelationship between the change of kisspetpin neurons and athe change of reproductive function with aging, 3) the mapping of kisspeptin receptor expression in the rat brain, and 4) the change of expression of kisspepin of certain kind of model animals suc as PCOS an/or hypothyroidism.

    researchmap

  • 多重プロモーターと選択的スプライシングによるエストロゲン受容体発現調節機構の解明

    2014

    科学研究費補助金 基盤研究(C) 

    石井 寛高

      More details

    Authorship:Principal investigator  Grant type:Competitive

    Grant amount:\1200000

    researchmap

  • 新規生殖制御系への環境情報入力と三次元的相互作用、生活環に関する機能形態学的研究

    2014

    科学研究費補助金 基盤研究(C) 

    小澤 一史, 飯島 典生, 託見 健, 石井 寛高, 岩田 衣世, 肥後 心平

      More details

    Authorship:Collaborating Investigator(s) (not designated on Grant-in-Aid)  Grant type:Competitive

    Grant amount:\1500000

    researchmap

  • Regulatory mechanisms of estrogen receptor gene expression by alternative promoter usage and alternative splicing

    Grant number:25460319  2013.4 - 2016.3

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research Grant-in-Aid for Scientific Research (C)  Grant-in-Aid for Scientific Research (C)

    Ishii Hirotaka

      More details

    Grant amount:\4940000 ( Direct Cost: \3800000 、 Indirect Cost:\1140000 )

    To determine regulatory mechanisms of estrogen receptor gene expression, alternative promoter usage and alternative splicing profiles of the estrogen receptor genes were examined. We identified modular structures of the estrogen receptor genes and several splice variants encoding N- and C-terminally-truncated estrogen receptor proteins. Some C-terminally-truncated estrogen receptor variants exhibited constitutive transcriptional transactivation in transfected cells. Ligand-independent transcriptional transactivation by steroid hormone receptors is deduced to be one of the candidate causes of malignant development of hormone-sensitive tumors. Therefore, our results may provide key mechanistic information explaining the cause of hormone-independent cell overgrowth in estrogen-sensitive tumors.

    researchmap

  • 多重プロモーターと選択的スプライシングによるエストロゲン受容体発現調節機構の解明

    2013

    科学研究費補助金 基盤研究(C) 

    石井 寛高

      More details

    Authorship:Principal investigator  Grant type:Competitive

    Grant amount:\1820000

    researchmap

  • 神経系における核外エストロゲン受容体を介したエストロゲン急性作用機構の解明

    2012

    科学研究費補助金 若手研究(B) 

    石井 寛高

      More details

    Authorship:Principal investigator  Grant type:Competitive

    Grant amount:\1040000

    researchmap

  • Brain mechanism mediating estrogen feedback controlling follicular development and ovulation

    Grant number:23380163  2011.4 - 2014.3

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research Grant-in-Aid for Scientific Research (B)  Grant-in-Aid for Scientific Research (B)

    TSUKAMURA Hiroko, ISHII Hirotaka, HIRABAYASHI Masumi

      More details

    Grant amount:\18330000 ( Direct Cost: \14100000 、 Indirect Cost:\4230000 )

    The present study aimed to investigate the brain mechanism mediating estrogen feedback to regulate two modes (pulse and surge) of gonadotropin-releasing hormone (GnRH)/gonadotropin release. Kisspeptin neurons located in the anteroventral periventricular nucleus (AVPV) and hypothalamic arcuate nucleus (ARC) are considered to control follicular development and ovulation via GnRH pulse and surge, respectively. In vivo and in vitro study using rats and mice revealed that epigenetic mechanism is involved in the both negative and positive feedback effects of estrogen to control expression of kisspeptin located in the AVPV and ARC. Since kisspeptin neurons are considered to govern reproductive function in mammals, the present results are expected to be applied to increase animal production efficiency in the future.

    researchmap

  • Ion Channels Related to Cell Excitability of GnRH Neurons

    Grant number:23590284  2011 - 2013

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research Grant-in-Aid for Scientific Research (C)  Grant-in-Aid for Scientific Research (C)

    KATO Masakatsu, ISHII Hirotaka, SAKUMA Yasuo

      More details

    Grant amount:\5200000 ( Direct Cost: \4000000 、 Indirect Cost:\1200000 )

    This study aimed to determine ion channels which closely related to the excitability of rat GnRH neurons. First, we analyzed K channels and found an involvement of somatostatin receptors (Sst), SK channels,and Kv7 channels. SK and Kv7 are involved in the generation of afterhyperpolarization. Second, cationic channels were analyzed in oligo-cell RT-PCR. GnRH neurons express Nalcn (Na leak channel), Unc-80 (associate protein to Nalc), and HCN (Hyperpolarization-activated cNucleotide gated Cationic channels).
    These ion channels may determine the cell excitability of GnRH neurons.

    researchmap

  • Expression profiles of G-protein coupled receptors in gonadotropin-releasing hormone neurons

    Grant number:23580402  2011 - 2013

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research Grant-in-Aid for Scientific Research (C)  Grant-in-Aid for Scientific Research (C)

    UENOYAMA Yoshihisa, OISHI Shinya, ISHII Hirotaka, SUWA Makiko

      More details

    Grant amount:\5330000 ( Direct Cost: \4100000 、 Indirect Cost:\1230000 )

    Transcriptome analysis with Sevens, a database of G-protein coupled receptors (GPCR), showed the expression profiles of GPCR in gonadotropin-releasing hormone (GnRH) neurons obtained from GnRH-green fluorescent protein transgenic mice. Among those GPCR, kisspeptin receptor GPR54 is considered as a key molecule controlling GnRH secretion. This study also showed brain region specific enhancer of Kiss1 gene and histone acetylation and chromatin loop formation are responsible for Kiss1 gene up-regulation.

    researchmap

  • 性ホルモンによる脳内神経回路の再構成が思春期に固有の社会行動を発現する機序の解明

    2011

    科学研究費補助金 基盤研究(B) 

    佐久間 康夫, 加藤 昌克, 木山 裕子, 折笠 千登世, 濱田 知宏, 石井 寛高

      More details

    Authorship:Collaborating Investigator(s) (not designated on Grant-in-Aid)  Grant type:Competitive

    Grant amount:\3500000

    researchmap

  • 神経系における核外エストロゲン受容体を介したエストロゲン急性作用機構の解明

    2011

    科学研究費補助金 若手研究(B) 

    石井 寛高

      More details

    Authorship:Principal investigator  Grant type:Competitive

    Grant amount:\800000

    researchmap

  • Molecular mechanisms involved in the site-specific regulation of estrogen receptor alpha expression in the brain and pituitary gland.

    2010.4 - 2011.3

    Japan Neuroendocrine Society  Young Investigator Grant of Japan Neuroendocrine Society 

    Hirotaka Ishii

      More details

    Authorship:Principal investigator 

    researchmap

  • Molecular mechanism for sex-specific reorganization of prosocial brain function during puberty

    Grant number:22390043  2010 - 2012

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research Grant-in-Aid for Scientific Research (B)  Grant-in-Aid for Scientific Research (B)

    SAKUMA Yasuo, KATO Masakatsu, KIYAMA Yuko, KONDO Yasuhiko, ORIKASA Chitose, HAMADA Tomohiro, HAMADA Tomohiro

      More details

    Grant amount:\18590000 ( Direct Cost: \14300000 、 Indirect Cost:\4290000 )

    In rodents, activation of estrogen receptor α (ERα) determines sexual phenotype of the brain a particular stage of ontogeny. Testosterone secreted by the testes during late gestational and neonatal periods is aromatized to form estradiol in the brain. Estradiol then masculinizes the brain through genomic activation of ERα; the lack of testosterone culminates in the female phenotype. The brain sexual phenotype determines sex-specific behavior and endocrinology in adults. We have shown that the sexual differentiation of the sexually dimorphic nucleus of the preoptic area (SDN-POA), which is larger in males than in females, is accomplished by estrogen-induced neuronal migration, by using a trait of transgenic rat. In the transgenics, neurons in the SDN-POA were labeled by fluorescent protein, EGFP. Migration was visualized by time-lapse microscopy of ex vivo slice culture of the brain. Further molecular biological experiments revealed the genomic activation of ERα culminates in pho phorylation/dephosphorylation kinetics of coffilin, which eventually regulates neuronal migration by altering actin dynamics. Recordings of neuronal activity in from this structure in the non-anesthetized, free-moving rat showed association of increased neuronal activity in males engaging in sexual interaction with females. Our observation of male-typical SDN-POA in oxytocin-ligand knock-out mice, which lacks mele-typical behavior, suggested the necessity of reassessment of the function of this structure in the future.

    researchmap

  • Characterization of extra-nuclear estrogen receptors in the central nervous system.

    Grant number:22790229  2010 - 2012

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research Grant-in-Aid for Young Scientists (B)  Grant-in-Aid for Young Scientists (B)

    ISHII Hirotaka

      More details

    Grant amount:\3250000 ( Direct Cost: \2500000 、 Indirect Cost:\750000 )

    This research was planned to characterize extra-nuclear estrogen receptors that mediate non-genomic effects of estrogens in the central nervous system. The genomic organization of human, mouse, and rat estrogen receptor α (ERα) genes was re-examined. The analysis revealed more complicated genomic structures of the ERα genes and localization of novel promoters and exons in the 5’-, intronic and 3’-regions of the genes. Furthermore, the analysis demonstrated that alternative promoter usage and alternative splicing generated numerous mRNA variants that encoded N-terminally- and C-terminally-truncated estrogen receptors. The variant proteins exhibited distinct functional properties and cellular localization compared with those of the wild-type ERα proteins.

    researchmap

  • 神経系における核外エストロゲン受容体を介したエストロゲン急性作用機構の解明

    2010

    科学研究費補助金 若手研究(B) 

    石井 寛高

      More details

    Authorship:Principal investigator  Grant type:Competitive

    Grant amount:\900000

    researchmap

  • 幼弱期の脳で合成される脳ニューロステロイドは神経回路構築を調節する

    2008

    科学研究費補助金 若手研究(B) 

    石井 寛高

      More details

    Authorship:Principal investigator  Grant type:Competitive

    Grant amount:\1500000

    researchmap

  • Brain neurosteroids synthesized in the neonatal brain modulate the construction of neural circuits.

    Grant number:19790181  2007 - 2008

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research Grant-in-Aid for Young Scientists (B)  Grant-in-Aid for Young Scientists (B)

    ISHII Hirotaka

      More details

    Grant amount:\3550000 ( Direct Cost: \3100000 、 Indirect Cost:\450000 )

    researchmap

  • 幼弱期の脳で合成される脳ニューロステロイドは神経回路構築を調節する

    2007

    科学研究費補助金 若手研究(B) 

    石井 寛高

      More details

    Authorship:Principal investigator  Grant type:Competitive

    Grant amount:\1600000

    researchmap

▼display all

Teaching Experience

  • Structure and function of the human body 2

    2024.4
    Institution:Nippon Medical School

     More details

  • Structure and function of the human body 1

    2024.1
    Institution:Nippon Medical School

     More details

  • 基礎医学総論I

    2023.3
    Institution:日本医科大学

     More details

  • 形態機能学Ⅱ

    2022.4
    Institution:日本医科大学専門学校

     More details

  • 形態機能学Ⅰ

    2022.3
    Institution:日本医科大学看護専門学校

     More details

  • 神経生物学

    2022.3
    Institution:国際基督教大学

     More details

  • 神経解剖学実習

    Institution:日本医科大学

     More details

  • 神経解剖学講義

    Institution:日本医科大学

     More details

  • 肉眼解剖学実習

    Institution:日本医科大学

     More details

  • 解剖生理学

     More details

  • 生理学実習

    Institution:日本医科大学

     More details

  • 呼吸生理学

    Institution:日本医科大学

     More details

▼display all