Updated on 2024/02/02

写真a

 
Hamada Tomohiro
 
Affiliation
Faculty of Medicine, Laboratory for Clinical Research, Collaborative Research Center, Assistant Professor
Title
Assistant Professor
External link

Research Interests

  • トランスジェニック

  • プロモーター

  • トランスジェニックラット

  • エストロジェン

  • 性差

  • 性的二型核

  • 視床下部腹内側核

  • エストロジェン受容体

  • 海馬

  • 性行動

  • 膜電位依存性チャネル

  • 電気生理学的解析

  • ロードシス

  • 視索前野

  • 視索前野性的二型核

  • 最後野

  • 電気生理学

  • 血圧調節

  • 電気生理

  • GFP

  • エストロジェン受容体遺伝子プロモーター

  • 遺伝子プロモーター

  • 性分化

  • カリウム電流

  • EGFP

  • ニューロステロイド

  • GABA

  • 内側視索前野

  • エストロゲン受容体

  • 免疫組織化学

Research Areas

  • Life Science / Neuroscience-general

Research History

  • Nippon Medical School   Medical School   Assistant Professor

    2010 - 2011

      More details

  • Nippon Medical School   Medical School   Assistant Professor

    2007 - 2008

      More details

  • Nippon Medical School   Medical School

    2000 - 2007

      More details

Professional Memberships

Papers

  • Estrogen-induced cell signaling in the sexually dimorphic nucleus of the rat preoptic area: Potential involvement of cofilin in actin dynamics for cell migration Reviewed

    Yuko Wada-Kiyama, Chiaki Suzuki, Tomohiro Hamada, Dilip Rai, Ryoiti Kiyama, Makoto Kaneda, Yasuo Sakuma

    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS   434 ( 2 )   287 - 292   2013.5

     More details

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

    Estrogen is a key factor to induce the sexually dimorphic nucleus (SDN) in the preoptic area (POA) of the rat brain. Identification of estrogen-dependent signaling pathways at SDN in POA during the critical period is a prerequisite for elucidating the mechanism. In the present study, we treated female rats with/without 17 beta-estradiol (E-2) at birth, designated as postnatal day I (P1), and prepared total RNA from brain slices containing SDN for DNA microarray analysis. Among the estrogen-responsive genes identified, protein kinase C-delta (PKC-delta) was significantly up-regulated by E-2 at P5. We examined the downstream effectors of PKC-delta protein by Western blotting and found an E-2-induced PKC-delta/Rac1/PAK1/LIMK1/cofilin pathway. In the pathway, E-2 suppressed the phosphorylation (inactive form) of cofilin. This result was supported by immunohistochemistry, where the phosphorylation/dephosphorylation of cofilin occurred at SDN, which suggests that cell migration is a cue to create sexual dimorphism in POA. (C) 2013 Elsevier Inc. All rights reserved.

    DOI: 10.1016/j.bbrc.2013.02.117

    Web of Science

    PubMed

    researchmap

  • Identification of C-terminally and N-terminally truncated estrogen receptor alpha variants in the mouse Reviewed

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

    JOURNAL OF STEROID BIOCHEMISTRY AND MOLECULAR BIOLOGY   124 ( 1-2 )   38 - 46   2011.3

     More details

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

    We re-examined mouse ER alpha mRNA variants using rapid amplification of cDNA ends (RACE) and RTPCR. 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 ER alpha products (CTERPs) were similar to that of Wild-type ER alpha and that the N-terminally truncated ER alpha. 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 ER alpha 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 ER alpha mRNAs and proteins. (C) 2011 Elsevier Ltd. All rights reserved.

    DOI: 10.1016/j.jsbmb.2011.01.003

    Web of Science

    PubMed

    researchmap

  • 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

  • Estrogen Receptor alpha Gene Promoter 0/B Usage in the Rat Sexually Dimorphic Nucleus of the Preoptic Area Reviewed

    Tomohiro Hamada, Yasuo Sakuma

    ENDOCRINOLOGY   151 ( 4 )   1923 - 1928   2010.4

     More details

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

    The volume of the sexually dimorphic nucleus of the preoptic area (SDN-POA) is two to four times larger in male rats than in females; however, the mechanism for the establishment of sexual dimorphism and the function of this nucleus is almost unknown. Perinatal estrogen can cause sexual dimorphism via the estrogen receptor alpha (ER alpha). Recently, transgenic rats were generated that express enhanced green fluorescent protein (EGFP) under the control of the ER alpha gene promoter 0/B to tag ER alpha-positive neurons in the brain. In the present study, we examined whether this EGFP expression could be a marker for the SDN-POA in adults. EGFP-labeled cells were distributed in the core of the SDN-POA (0/B-SDN)of male and female transgenic rats, in accordance with the Nissl staining and immunoreactivity for the SDN marker, calbindin. They were also immunoreactive for ER alpha. The core was bigger in volume and contained more 0/B-SDN neurons in males than in females. The EGFP-tagged cells were packed more densely in the female core than that in males. Subcutaneous injection of 100 mu g 17 beta-estradiol to females on the day of birth, or orchidectomy of male neonates, reversed the sexually dimorphic phenotype of the volume of the 0/B-SDN, despite not affecting the cell number. We suggest that this EGFP expression in the SDN-POA could be a useful marker to clarify the sexual differentiation and function of the SDN-POA. Moreover, the ER alpha gene promoter 0/B plays a key role in the organization of the sexual differentiation of the SDN-POA. ( Endocrinology 151: 1923-1928, 2010)

    DOI: 10.1210/en.2009-1022

    Web of Science

    PubMed

    researchmap

  • In vivo Visualization of the Rat Sexually Dimorphic Nucleus of the Preoptic Area

    Hamada Tomohiro, Sakuma Yasuo

    Nihon Ika Daigaku Igakkai Zasshi   6 ( 3 )   96 - 97   2010

     More details

    Language:Japanese   Publisher:The Medical Association of Nippon Medical School  

    DOI: 10.1272/manms.6.96

    researchmap

  • Site-specific regulation of gene expression by estrogen in the hypothalamus of adult female rats. Reviewed International journal

    Xu Q, Hamada T, Kiyama R, Sakuma Y, Wada-Kiyama Y

    Neuroscience letters   436 ( 1 )   35 - 39   2008.5

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:1  

    Estrogen plays critical roles in the neuroendocrine system of adult female rats through separate actions, respectively, in the preoptic area (POA) and the ventromedial nucleus of the hypothalamus (VMH). Seven-week-old rats were treated with/without estrogen after they were ovariectomized, and four estrogen-responsive, neuronal system-related genes, encoding alpha4 neuronal nicotinic acetylcholine receptor (Chrna4), GABA(A) receptor delta (Gabrd), serotonin receptor 6 (Htr6), and GABA transporter 2 (Slc6a13), were investigated by real-time RT-PCR and Western blot analyses to examine their differential regulation by estrogen between the anterior part containing POA and the posterior part containing VMH. We further examined Bax, Bcl2, and Prkce, the former two genes to be involved in the gene expression network of Chrna4 and the latter gene, that of Gabrd. The regulation of Bax and Bcl2 by estrogen differed between the anterior and posterior parts. The results demonstrated differential regulation of these neuronal system-related genes by estrogen between the anterior and posterior parts of the hypothalamus and suggested the roles of gene expression networks for the respective genes in the neuroendocrine system of adult female rats.

    DOI: 10.1016/j.neulet.2008.02.054

    PubMed

    researchmap

  • In vivo visualization of estrogen receptor α gene promoter activity Reviewed

    Tomohiro Hamada, Yasuo Sakuma

    Journal of Nippon Medical School   73 ( 3 )   114 - 115   2006

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:3  

    DOI: 10.1272/jnms.73.114

    Scopus

    PubMed

    researchmap

  • Visualizing forebrain-specific usage of an estrogen receptor at promoter for receptor downregulation in the rat Reviewed

    T Hamada, Y Wada-Kiyama, Y Sakuma

    MOLECULAR BRAIN RESEARCH   139 ( 1 )   42 - 51   2005.9

     More details

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

    Transgenic rats expressing enhanced green fluorescent protein (EGFP) under the control of an estrogen receptor (ER) alpha promoter were generated to tag ER alpha-positive neurons in the brain. Two transgenes, one containing sequences for promoter A and DsRed and the other containing sequences for promoter 0/B and EGFP, were injected simultaneously into Wistar rat zygotes. Twenty-two founders with both transgenes were identified. Ten lines of these founders expressed the EGFP tag in the brains of their first filial generation, whereas none similarly expressed the DsRed tag. In two lines selected for the brightness of the EGFP fluorescence in their brains, tagged cells showed essentially the same patterns. Tagged cells were in the preoptic area (POA), bed nucleus of the stria terminalis (BNST), hypothalamic arcuate nucleus and medial amygdala. ER alpha-immunoreactive neurons were identified in all of these structures by immunohistochemistry. In ovariectomized females, approximately 75% of the EGFP-fluorescent cells in the POA-BNST were immunoreactive for ER alpha. In the POA-BNST, ovariectomy increased the number of EGFP-immunopositive cells and estrogen supplementation reversed this effect, indicating that the promoter 0/B is involved in estrogen-induced downregulation of ER alpha. EGFP was also present in cells in the cerebral cortex and hippocampus, which have not previously been associated with endocrine regulation. Conversely, only a few cells were tagged in the hypothalamic ventromedial nucleus, which contained many ER alpha-immunoreactive neurons. This discrepancy could have arisen as a result of differential promoter usage. (c) 2005 Elsevier B.V. All rights reserved.

    DOI: 10.1016/j.molbrainres.2005.05.019

    Web of Science

    PubMed

    researchmap

  • Single-cell real-time quantitative polymerase chain reaction of immunofluorescently identified neurons of gonadotropin-releasing hormone subtypes in cichlid fish Reviewed

    IS Parhar, S Ogawa, T Hamada, Y Sakuma

    ENDOCRINOLOGY   144 ( 8 )   3297 - 3300   2003.8

     More details

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

    The regulatory mechanisms controlling gene expression of GnRH subtypes, particularly of the evolutionarily conserved GnRH2, remain speculative. To address this issue, we have successfully coupled the anatomic specificity of immunofluorescently defined "cell picking" with the sensitivity of real-time quantitative RT-PCR (RT-Q-RT-PCR), which enabled us to examine the presence and quantity of GnRH mRNAs in individual neurons. Here, using RT-Q-RT-PCR, we report change in the levels of transcripts of GnRH subtypes in individual neurons harvested from the brain of mature and immature males of tilapia, Oreochromis niloticus. The levels of GnRH1mRNA per cell and the percentage of neurons expressing GnRH1 transcripts exceeding 0.05 x 10(2) fg/cell were significantly higher in mature males (44.2%) compared with immature males (4.7%). In contrast, there was no difference in mRNA levels and the percentage of cells expressing GnRH2 and GnRH3 between the two reproductive states. Thus, using a novel approach that enables immunofluorescently labeled single-cell RT-Q-RT-PCR analysis of GnRH neurons, we present evidence that shows preoptic GnRH1 is important for gonadal maturation, whereas GnRH2 and GnRH3 might have supplementary roles in reproductive behaviors or nonreproductive functions. Furthermore, we speculate that the use of this method will allow the identification and quantification of known and unknown genes in single GnRH neurons, which would greatly facilitate our understanding of the complex interactions that govern the physiology of individual cells of GnRH variants in vertebrate species.

    DOI: 10.1210/en.2003-0386

    Web of Science

    PubMed

    researchmap

▼display all

Books

  • Adaptation Biology and Medicine Vol.8

    Tomohiro Hamada(Sexual dimorphism in the preoptic area: An adaptive outcome to environment.)

    Narosa Publishing House  2017 

     More details

  • カラー図解症状の基礎からわかる病態生理

    濱田知宏( Role: Joint translatorホルモン)

    メディカル・サイエンス・インターナショナル  2011.9 

     More details

Misc.

  • 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

    日本内分泌学会雑誌   87 ( 2 )   617 - 617   2011.9

     More details

    Language:Japanese   Publisher:(一社)日本内分泌学会  

    J-GLOBAL

    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

    日本下垂体研究会学術集会プログラム・講演要旨集   26th   31   2011

     More details

    Language:Japanese  

    J-GLOBAL

    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

    日本下垂体研究会学術集会プログラム・講演要旨集   26th   31   2011

     More details

    Language:Japanese  

    J-GLOBAL

    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。

    石井寛高, 庄田有里, 濱田知宏, 佐久間康夫

    日本内分泌学会雑誌   86 ( 2 )   344 - 344   2010.9

     More details

    Language:Japanese   Publisher:(一社)日本内分泌学会  

    J-GLOBAL

    researchmap

  • Estrogen-responsive genes in the preoptic area of the female rat during the critical period for sexual differentiation of the brain

    Yuko Wada-Kiyama, Chiaki Suzuki, Tomohiro Hamada, Ryoiti Kiyama, Yasuo Sakuma

    JOURNAL OF PHYSIOLOGICAL SCIENCES   60   S174 - S174   2010

     More details

    Language:English   Publishing type:Research paper, summary (international conference)   Publisher:SPRINGER TOKYO  

    Web of Science

    researchmap

  • Neuronal migration is important for the sexual differentiation of the sexually dimorphic nucleus of the preoptic area

    Tomohiro Hamada, Yasuo Sakuma

    JOURNAL OF PHYSIOLOGICAL SCIENCES   60   S175 - S175   2010

     More details

    Language:English   Publishing type:Research paper, summary (international conference)   Publisher:SPRINGER TOKYO  

    Web of Science

    researchmap

  • VISUALIZATION OF NUCLEOGENESIS OF THE SEXUALLY DIMORPHIC NUCLEUS OF THE PREOPTIC AREA

    Tomohiro Hamada, Yasuo Sakuma

    JOURNAL OF PHYSIOLOGICAL SCIENCES   59   229 - 229   2009

     More details

    Language:English   Publishing type:Research paper, summary (international conference)   Publisher:SPRINGER TOKYO  

    Web of Science

    researchmap

  • The efferent connections of the sexually dimorphic nucleus in the preoptic area

    Tomohiro Hamada, Chunxiao Zhang, Yasuo Sakuma

    NEUROSCIENCE RESEARCH   61   S109 - S109   2008

     More details

    Language:English   Publishing type:Research paper, summary (international conference)   Publisher:ELSEVIER IRELAND LTD  

    Web of Science

    researchmap

  • Estrogen modulates voltage dependent potassium currents in the EGFP-tagged neurons in the hippocampus of estrogen receptor gene promoter transgenic rats

    Tomohiro Hamada, Hongli Shan, Yasuo Sakuma

    NEUROSCIENCE RESEARCH   58   S186 - S186   2007

     More details

    Language:English   Publishing type:Research paper, summary (international conference)   Publisher:ELSEVIER IRELAND LTD  

    Web of Science

    researchmap

  • Estrogen receptor a gene promoter activity is a marker for the sexually dimorphic nucleus of the preoptic area

    Tomohiro Hamada, Yasuo Sakuma

    NEUROSCIENCE RESEARCH   55   S158 - S158   2006

     More details

    Language:English   Publishing type:Research paper, summary (international conference)   Publisher:ELSEVIER IRELAND LTD  

    Web of Science

    researchmap

▼display all

Presentations

  • 脳の性分化機構に置けるエストロゲンの細胞移動調節 Invited

    濱田知宏

    第17回早稲田大学「性と生殖」公開シンポジウム  2011.12 

     More details

    Language:Japanese   Presentation type:Oral presentation (invited, special)  

    researchmap

Research Projects

  • 心の性決定神経回路形成を主導する思春期エストロゲン

    2018.4 - 2021.3

    文部科学省  科学研究費助成事業(基盤研究C) 

    濱田知宏

      More details

    Authorship:Principal investigator  Grant type:Competitive

    researchmap

  • Sexually dimorphic forebrain nucleus and social bonding

    Grant number:17K08578  2017.4 - 2020.3

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

    Sakuma Yasuo

      More details

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

    Oxytocin (oxt) receptors in the preoptic area (POA), which include hormone-sensitive sexually dimorphic nuclus (SDN-POA), have been implicated in sexual partner preference in males in various species. In male mice with oxt knockout, preference to estrous females was lost, however, the size of the SDN-POA remained as same as in wild-type males. The observation contradicted the prevailing notion and called for a revision. In the current series of experiments, we showed (1) oxt knock-outs diminished sexual partner preference in both male and female mice; (2) a lacrimal protein in rats, cystatin-related protein 1, induces defensive response through the vomeronasal system, a major source of neural inputs to the POA; (3) ticking of the young rats, which replicated cutaneous sensory stimulation during rough-and-tumble play of the animal, established avid attachment of the animal to the experimenter: the animal would be useful in the animal assisted therapy in rehabilitation medicine.

    researchmap

  • Sexual differentiation of the sexually dimorphic nucleus of the preoptic area and the bed nucleus of the stria terminalis responsible for the gender identity.

    Grant number:25460320  2013 - 2016

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

    Hamada Tomohiro

      More details

    Authorship:Principal investigator  Grant type:Competitive

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

    Sex differences of the brain would be important for the gender identity, however, the mechanism of the sexual differentiation of the brain remains largely unknown. The sexually dimorphic nucleus of the preoptic area (SDN-POA) is thought to contribute to the gender identity. In this study, we examined the sexual differentiation of the SDN-POA using the organotypic brain slice cultures and time-lapse imaging in vitro to test the hypothesis that neural migration regulated by estrogen is critical for the sexual differentiation of the SDN-POA. The scattering neural migration by estrogen was observed in the masculinization of the SDN-POA. This estrogen induced scattering migration was mediated through the estrogen receptor alpha/Rac1/cofilin/actin pathway. These results suggest that scattering neural migration by estrogen is critical role for the masculinization of the SDN-POA.

    researchmap

  • Visualization of the sexual differentiation in the sexually dimorphic nucleus of the preoptic area.

    Grant number:22500297  2010 - 2012

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

    HAMADA Tomohiro, SAKUMA Yasuo

      More details

    Authorship:Principal investigator  Grant type:Competitive

    Grant amount:\3510000 ( Direct Cost: \2700000 、 Indirect Cost:\810000 )

    The sexually dimorphic nucleus in the preoptic area (SDN-POA) is larger in males than in females, however the mechanism of sexual differentiation of this structure remains largely unknown, except that androgen and/or estrogen during perinatal period cause the establishment of male-typical nuclei. We have shown recently that enhanced green fluorescent protein (EGFP) is expressed in the SDN-POA under the control of an estrogen receptor α gene promoter. The present study suggested that neural migration plays a key role in the sexual differentiation of the SDN-POA to visualize the nucleogenesis of the SDN-POA in vivo and in vitro.

    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)

    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

  • Sex steroids and the sexually dimorphic nucleus of the rat preoptic area

    Grant number:20590238  2008 - 2010

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

    ORIKASA Chiotse, SAKUMA Yasuo, HAMADA Tomohiro

      More details

    Grant amount:\4810000 ( Direct Cost: \3700000 、 Indirect Cost:\1110000 )

    Although the SDN-POA was significantly larger in males than in females at PD15, the total numbers of neurons comprising the SDN-POA were not significantly different between sexes. Sexual dimorphism in the SDN-POA results from male-specific postnatal radial spreading of cells rather than cell proliferation during embryonic neurogenesis.

    researchmap

  • INTRACELLULAR ION ENVIRONMENT DURING THE ONTOGENY AS A DETERMINANT OF SEXUAL PHENOTYPE OF RAT BRAIN

    Grant number:18390070  2006 - 2009

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

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

      More details

    Grant amount:\17450000 ( Direct Cost: \14300000 、 Indirect Cost:\3150000 )

    In the rat, estrogen determines sexual phenotype of the brain, acting at a critical period as neonates. The sexual phenotype thus established persists into adulthood, both in morphology and electrophysiology. In the adults, the volume of the sexually dimorphic nucleus of the preoptic area (SDN-POA) is larger in males than in females. The anteroventral periventricular nucleus (AVPV) is packed with estrogen receptor (ER) ss-positive neurons in females but these cells scatter in the more lateral areas in males. Estrogen regulates axonal excitability of projection neurons in the ventromedial hypothalamus and those in the preoptic area in diametrically opposite directions in females but not in males. Male phenotype depends on the presence of estrogen as neonates ; the lack leads to the female phenotype. This research project focused intracellular signal cascades, which are kindled by estrogen through estrogen receptor (ER) α and culminate in neurogenesis, migration or cell death. 1) By using 5'-RACE we identified a new leader exon and untranslated internal exons in ERα gene. These exons were used for site-specific transcription of ERα ; 2) In a trait of transgenic rat, which express EGFP under the control of ERα promoter 0/B, ERα-positive neurons in the SDN-POA, but not those in the adjacent areas, were fluorescent. Time lapse movies showed the establishment of the SDN-POA as a result of neuronal migration ; 3) In another trait of transgenic rat, in which gonadotropin-releasing hormone (GnRH) neurons were tagged by EGFP, activation of GABAA_A receptors depolarized adult GnRH neurons due to their high chloride ion content ; 4) DNA microarray, PCR and Western-blot analysis demonstrated site-specific, opposite regulations of apoptosis- and migration-related genes in the SDN-POA and AVPV.

    researchmap

  • 神経活動依存的逆行性標識による性腺刺激ホルモン排卵性大量分泌中枢の特定

    Grant number:18659061  2006 - 2007

    日本学術振興会  科学研究費助成事業  萌芽研究

    佐久間 康夫, 木山 裕子, 濱田 知宏

      More details

    Grant amount:\3300000 ( Direct Cost: \3300000 )

    性腺刺激ホルモン放出ホルモン(GnRH)ニューロンは脳による生殖内分泌調節の最終共通路であり、上位の脳機構からの支配を受けて、下垂体前葉を調節し性腺からのホルモン分泌調節や配偶子の形成を行っている。上位の脳機構についてはほとんどわかっていない。γ-アミノ酪酸(GABA)A受容体(GABA_AR)を介するGABA、AMPA受容体を介するグルタミン酸、キスペプチンなどのペプチドがGnRHニューロンの活動を調節すると考えられている。本研究では当初、古典的経シナプス性制御に着目し、無毒化した破傷風毒素(TTC)が神経活動依存的に逆行性系シナプス性標識を行うという特徴を活用し、GnRHニューロンに投射しているニューロンをトランスジェニックラットで可視化することを試みた。GnRHプロモーターの下流に蛍光蛋白であるEGFPとTTC遺伝子をつなげた導入遺伝子を用い、4系統のトランスジェニックラットを得、サザンブロット法によりこれらのラットに遺伝子導入が起こっていることを確認し表現型を検討したが、何れの系統においても脳内GnRHニューロンあるいは他のニューロンにEGFP標識が見られなかった。性腺摘除を行ってフィードバック環を開放してGnRHニューロンの過剰な活動を起こしたり、経代を重ねることで目的の表現型が得られるかについても検討したが、計画年度内には成功に至らなかった。一方、蛍光タンパク遣伝子の導入により、可視化したラットGnRHニューロンを対象とする実験では、GnRHニューロンにGABA_ARのα2,β3,γ1またはγ2サブユニットが発現していることをRT-PCR法で確認し、グラミシジン穿孔パッチクランプ法により、GnRHニューロンでは細胞内塩素イオン濃度が高く、成熟後もGABA_ARの活性化が脱分極を起こすこと、低濃度のGABAは活動電位の発生を促すが、高濃度では脱分極ブロックにより、活動電位の発生を抑えることを見いだし報告した(Yin etal.,2008)。gabazineによるGABA_A電流の阻止効果が限定的であったこと、この実験における低濃度のGABAは前脳底部におけるシナプス外GABAの濃度に相当することの2点から、シナプス外のGABA_ARの活性化がGnRHニューロンの調節に大きな役割を果たしていることが示唆され、本実験計画の当初の仮説の妥当性を考え直す契機となった。以上、本研究計画は当初の成果を挙げられなかったが、GnRHニューロン、ひいては視床下部ペプチド作動性ニューロンの調節一般について、古典的考え方にとらわれない新規な発想を導くに至った点で、有益であった。

    researchmap

  • Elucidation of the multifunctional signaling of estrogen using the visualization of estrogen responsible neurons

    Grant number:17500215  2005 - 2007

    Ministry of Education, Culture, Sports, Science and Technology  Grants-in-Aid for Scientific Research(基盤研究(C))  基盤研究(C)

    Tomohiro HAMADA, Yasuo SAKUMA, Yuko KIYAMA

      More details

    Authorship:Principal investigator  Grant type:Competitive

    Grant amount:\3710000 ( Direct Cost: \3500000 、 Indirect Cost:\210000 )

    Estrogen plays an essential role for reproduction, plasticity and neuroprotection to affect on the central nervous system. The purpose of the present study is to examine the mechanisms of estrogen action using the visualized neurons that are responsible to estrogen in the estrogen receptor gene promoter transgenic rats. Most important result is that EGFP expression under the control of estrogen receptor gene promoter is a specific marker for the sexually dimorphic nucleus of the preoptic area. Because sexual dimorphism of this nucleus is established by estrogen during the perinatal period, the new, live marker would contribute to clarify the molecular mechanisms of the sexual differentiation. On the other hand, EGFP expression is observed in the hippocampus that is noted function of estrogen for the plasticity and neurogenesis. We determined acute and chronic effects of physiological concentration of estrogen on the outward potassium currents in the primary cultured EGFP hippocampal neurons. Immature neurons showed the facilitated effect of 48 h treatment of estrogen, however, mature neurons exhibited the suppressive effect on the outward potassium currents. After the 48 h treatment of estrogen, bath application of 250 pM estrogen enhanced the potassium currents. These results suggest that estrogen influence the development and excitability of hippocampal neurons. One more thing to be noted is that there is the sex difference in the number of EGFP expressed cells in the area postrema and the nucleus tractus solitarius that are known to play an important role for cardiovascular regulation. Because hypertension is inhibited by estrogen in female, we could expect to clear the mechanisms of estrogen function on the cardiovascular regulation. Thus, we can show a number of the important results and we would like to study further to make clear the molecular mechanisms of estrogen function in the central nervous system.

    researchmap

  • Identification of Molecular Mechanisms of Estrogen-Regulated Brain Sex Differentiation

    Grant number:16086210  2004 - 2008

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research  Grant-in-Aid for Scientific Research on Priority Areas

    SAKUMA Yasuo, ISHWAR S Parhar

      More details

    Grant amount:\120000000 ( Direct Cost: \120000000 )

    researchmap

  • 逆行性標識タンパク質特異的発現を利用したGnRH分泌調節回路の可視化

    Grant number:16659061  2004 - 2005

    日本学術振興会  科学研究費助成事業  萌芽研究

    佐久間 康夫, 加藤 昌克, 木山 裕子, 濱田 知宏

      More details

    Grant amount:\3200000 ( Direct Cost: \3200000 )

    視床下部に散在している性腺刺激ホルモン放出ホルモン(GnRH)ニューロンは、生殖内分泌系を調節する最終共通路であるが、支配する神経回路についてはほとんどわかっていない。本研究では、トランスジェニックラットにおいて逆行性トレーサーをGnRHニューロン特異的に発現させ、経ナプス的に支配ニューロンを可視化することを目論んだ。すなわち蛍光タンパク質(EGFP)と無毒化テタヌストキシンC末端(TTC)の融合タンパク質を逆行性トレーサーとして用いた。前年度に作製したGnRHプロモーター-EGFP-TTCという導入遺伝子をラット受精卵に注入し、4系統のファウンダーラットを得た。サザンブロットの結果から導入された遺伝子は1コピー(1)、100コピー(2)および300コピー(1)と推定され、これまでの当研究室でのトランスジェニックラット作出の経験から、表現型発現が期待されるレベル(100-300)にあることがわかった。野生型と交配しそれぞれ産仔を得たが、ある系統では産仔に導入遺伝子がまったく伝達されなかった。また別の系統では雌のみに導入遺伝子が伝達され、このファウンダーが雄だったことから、導入遺伝子がX染色体上にあることが示唆された。他の2系統はメンデルの法則に則って産仔が得られた。これら産仔(4週令と7週令)の脳切片を作成し、EGFP蛍光発現を検討したが、どの系統も蛍光は観察されなかった。発現したEGFP分子が蛍光として可視されるほど多くないことを予想してGFPに対する兔疫染色を試したが、これもまったく染色されなかった。
    これらの結果は遺伝子導入実現が必ずしも表現型として表出されるとは限らないというトランスジェニックの宿命のためであると考えられる。現在更に系代し、表現型の確認を進めるとともに、導入遺伝子の更なる受精卵移植を検討しているところである。

    researchmap

  • Cellular physiological bases of sex-specific estrogen actions on the brain

    Grant number:14370025  2002 - 2005

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

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

      More details

    Grant amount:\14600000 ( Direct Cost: \14600000 )

    Transgenic rats expressing enhanced green fluorescent protein (EGFP) under the control of an estrogen receptor (ER) alpha promoter were generated to tag ERalpha-positive neurons in the brain. Two transgenes, one containing sequences for promoter A and DsRed and the other containing sequences for promoter 0/B and EGFP, were injected simultaneously into Wistar rat zygotes. Twenty-two founders with both transgenes were identified. Ten lines of these founders expressed the EGFP tag in the brains of their first filial generation, whereas none similarly expressed the DsRed tag. In two lines selected for the brightness of the EGFP fluorescence in their brains, tagged cells showed essentially the same patterns. Tagged cells were in the preoptic area (POA), bed nucleus of the stria terminalis (BNST), hypothalamic arcuate nucleus and medial amygdala. ERalpha-immunoreactive neurons were identified in all of these structures by immunohistochemistry. In ovariectomized females, approximately 75% of the EGFP-fluorescent cells in the POA-BNST were immunoreactive for ERalpha. In the POA-BNST, ovariectomy increased the number of EGEP-immunopositive cells and estrogen supplementation reversed this effect, indicating that the promoter 0/B is involved in estrogen-induced downregulation of ERalpha. EGFP was also present in cells in the cerebral cortex and hippocampus, which have not previously been associated with endocrine regulation. Conversely, only a few cells were tagged in the hypothalamic ventromedial nucleus, which contained many ERalpha-immunoreactive neurons. This discrepancy could have arisen as a result of differential promoter usage.

    researchmap

  • 雌性行動神経回路を構成し可視化した視床下部ニューロンに対するエストロジェンの作用

    Grant number:14780606  2002 - 2004

    文部科学省  科学研究費補助金(若手研究(B))  若手研究(B)

    濱田 知宏

      More details

    Authorship:Principal investigator  Grant type:Competitive

    Grant amount:\3100000 ( Direct Cost: \3100000 )

    エストロジエン受容体(ER)トランスジェニック(TG)ラットを用いた検討これまでの研究により、α型ERを発現するニューロンを生きた状態で可視化されるTGラットの作製に成功した。すなわち、ラットα型ER遺伝子プロモーターのひとつであるプロモーターO/Bの下流に、GFP遺伝子を結合させた導入遺伝子を用いることで、雌型性行動に対し抑制的に働く視索前野にGFPを発現するTGラットを得た。視索前野に存在するGFP発現細胞の70%は免疫組織学的にERを発現し、このGFP発現はERと同様にエストロジェンによってdown-regulateされていることから、プロモーターO/BがERのdown-regulationに関わっていることが明らかとなった(論文投稿中)。複数存在するER遺伝子プロモーターの機能的差異についてはほとんどわかっておらず、本研究の結果によりその一端が明らかとなった。また同時にER遺伝子プロモーターの制御下で発現したGFP蛍光を指標として、これまで困難であったER発現ニューロンを生きた状態で可視化することが可能となった。これら視索前野GFP発現細胞を用いた、エストロジェン作用に関する電気生理学的な研究を現在進めている。一方で雌型性行動に対し促進中枢と考えられ、BRの局在が観察される視床下部腹内側核にはほとんどGFPは発現していなかった。このことは視索前野と腹内側核で機能しているER遺伝子プロモーターが異なることを示唆している。以上のように雌型行動神経回路を可視化する目的で作製したTGラットを用いることで、本来の目的を達しただけでなく、これまで不明であった多種類ER遺伝子プロモーターの存在意義について重要な知見が得られた。特に雌の性行動に対し拮抗的に働いている二つの神経核で、異なるプロモーターが機能しているということは興味深く、今後の研究進展が期待される。

    researchmap

  • エストロジェンによるロードシス発現に伴う視床下部ニューロン興奮特性の変化

    Grant number:12770034  2000 - 2001

    文部科学省  科学研究費補助金(奨励研究(A))  奨励研究(A)

    濱田 知宏

      More details

    Authorship:Principal investigator  Grant type:Competitive

    Grant amount:\2000000 ( Direct Cost: \2000000 )

    発情期のげっ歯類の雌に見られるエストラジオール(E_2)依存性のロードシス発現には、中脳灰白質(PAG)に投射している視床下部腹内側核(VMH)のE_2感受性ニューロンが重要な役割を演じている。このニューロンに関する細胞レベルでの生理学的特性を明らかにするために、前年度の結果(投稿準備中)より、後部PAGの腹側に逆行性トレーサーを注入することで、生きた状態で蛍光ラベルされたVMHニューロンについてslice patch clamp法を用いた電気生理学的な検討を行った。まず視床下部で重要なトランスミッターであるGABAに対する反応性について検討を試みたが、細胞が非常に脆弱で、あまりデータが得られなかった。この原因は同条件で視索前野ニューロンについて記録可能であるためVMHニューロンの性質に起因すると考えられるが、一方で蛍光トレーサーの副作用である可能性も考えられる。現在、視床下部スライス作製時の条件の再検討を行っている。また実験計画には記載されていなかったが、E2感受性ニューロン、さらに言えばエストロジェン受容体を含有するニューロンを生きた状態で可視化する目的で、α型ER遺伝子のプロモーターに蛍光タンパクであるGFPを結合させたトランスジェニックラットを作製した。GFPの蛍光は脳内で確認され、視床下部のみならず、大脳皮質や海馬にもGFPでラベルされたニューロンが存在しており、免疫組織化学的にα型ERとの共存も確認された。今後はこのトランスジェニックラットのPAGに逆行性トレーサーを注入し、GFPと二重ラベルされた細胞について膜特性、特にエストロジェンの作用に着目して電気生理学的な研究を進めて行くつもりである。

    researchmap

▼display all