Network


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

Hotspot


Dive into the research topics where Shin-ichiro Honda is active.

Publication


Featured researches published by Shin-ichiro Honda.


Cell | 2009

Estrogen Masculinizes Neural Pathways and Sex-Specific Behaviors

Melody V. Wu; Devanand S. Manoli; Eleanor J. Fraser; Jennifer K. Coats; Jessica Tollkuhn; Shin-ichiro Honda; Nobuhiro Harada; Nirao M. Shah

Sex hormones are essential for neural circuit development and sex-specific behaviors. Male behaviors require both testosterone and estrogen, but it is unclear how the two hormonal pathways intersect. Circulating testosterone activates the androgen receptor (AR) and is also converted into estrogen in the brain via aromatase. We demonstrate extensive sexual dimorphism in the number and projections of aromatase-expressing neurons. The masculinization of these cells is independent of AR but can be induced in females by either testosterone or estrogen, indicating a role for aromatase in sexual differentiation of these neurons. We provide evidence suggesting that aromatase is also important in activating male-specific aggression and urine marking because these behaviors can be elicited by testosterone in males mutant for AR and in females subjected to neonatal estrogen exposure. Our results suggest that aromatization of testosterone into estrogen is important for the development and activation of neural circuits that control male territorial behaviors.


Hormones and Behavior | 2002

Sexual Partner Preference Requires a Functional Aromatase (Cyp19) Gene in Male Mice

Julie Bakker; Shin-ichiro Honda; Nobuhiro Harada; Jacques Balthazart

Sexual motivation, sexual partner preference, and sexual performance represent three different aspects of sexual behavior that are critical in determining the reproductive success of a species. Although the display of sexual behavior is under strict hormonal control in both sexes, the relative roles of androgen and estrogen receptors in activating the various components of male sexual behavior are still largely unknown. A recently developed mouse model that is deficient in estradiol due to targeted disruption of exons 1 and 2 of the Cyp19 gene (aromatase knockout (ArKO) mice) was used here to analyze the role of estradiol in the control of all three aspects of male sexual behavior. When tested in a Y-maze providing volatile olfactory cues, male ArKO mice did not show a preference for the odors from an estrous female over those from an intact male, whereas wild-type (WT) and heterozygous (HET) males clearly preferred to sniff estrous odors. When provided with visual and olfactory cues, male ArKO mice also failed to show a preference for an estrous female when given a choice between an estrous female and an empty arm. However, sexual partner preferences of male ArKO mice were not sex-reversed: they did not prefer to investigate an intact male over an estrous female or empty arm. Thus, male ArKO mice seemed to have general deficits in discriminating between conspecifics by using olfactory and visual cues. Male coital behavior was also severely impaired in male ArKO mice: they displayed significantly fewer mounts, intromissions, and ejaculations than WT and HET males. Latencies to first mount or intromission were also significantly longer in ArKO males compared to WT and HET males, in addition to them showing less interest in investigating olfactory and visual cues in a Y-maze, suggesting that they were sexually less motivated. However, three out of seven male ArKO mice were capable of siring litters provided they were housed with a female for a prolonged period of time. In conclusion, aromatization of testosterone to estradiol appears to be essential for sexual motivation and sexual partner preference. By contrast, estradiol may play only a limited role in the expression of male coital behaviors.


Neuron | 2010

The androgen receptor governs the execution, but not programming, of male sexual and territorial behaviors

Scott A. Juntti; Jessica Tollkuhn; Melody V. Wu; Eleanor J. Fraser; Stella Tan; Shin-ichiro Honda; Nobuhiro Harada; Nirao M. Shah

Testosterone and estrogen are essential for male behaviors in vertebrates. How these two signaling pathways interact to control masculinization of the brain and behavior remains to be established. Circulating testosterone activates the androgen receptor (AR) and also serves as the source of estrogen in the brain. We have used a genetic strategy to delete AR specifically in the mouse nervous system. This approach permits us to determine the function of AR in sexually dimorphic behaviors in males while maintaining circulating testosterone levels within the normal range. We find that AR mutant males exhibit masculine sexual and territorial displays, but they have striking deficits in specific components of these behaviors. Taken together with the surprisingly limited expression of AR in the developing brain, our findings indicate that testosterone acts as a precursor to estrogen to masculinize the brain and behavior, and signals via AR to control the levels of male behavioral displays.


Hormones and Behavior | 2004

Restoration of male sexual behavior by adult exogenous estrogens in male aromatase knockout mice

Julie Bakker; Shin-ichiro Honda; Nobuhiro Harada; Jacques Balthazart

We previously found that male aromatase knockout (ArKO) mice that carry a targeted mutation in exons 1 and 2 of the CYP19 gene and as a result cannot aromatize androgen to estrogen show impaired sexual behavior in adulthood. To determine whether this impairment was due to a lack of activation of sexual behavior by estradiol, we studied here male coital behavior as well as olfactory investigation of sexually relevant odors in male ArKO mice following adult treatment with estradiol benzoate (EB) or dihydrotestosterone propionate (DHTP). Again, we found that gonadally intact ArKO males show pronounced behavioral deficits affecting their male coital behavior as well as their olfactory investigation of volatile body odors but not that of soiled bedding. Deficits in male coital behavior were largely corrected following adult treatment with EB and the androgen DHTP, suggesting that estradiol has prominent activational effects on this behavior. By contrast, adult treatment with EB to either castrated or gonadally intact ArKO males did not stimulate olfactory investigation of volatile body odors, suggesting that this impairment may result from a lack of proper organization of this behavior during ontogeny due to the chronic lack of estrogens. In conclusion, the present studies suggest that the behavioral deficits in sexual behavior in male ArKO mice result predominantly from a lack of activation of the behavior by estrogens. This is in contrast with earlier pharmacological studies performed on rats and ferrets that have suggested strong organizational effects of estradiol on male sexual behavior.


The Journal of Physiology | 2008

Acute effects of oestrogen on the guinea pig and human IKr channels and drug-induced prolongation of cardiac repolarization

Junko Kurokawa; Masaji Tamagawa; Nobuhiro Harada; Shin-ichiro Honda; Chang-Xi Bai; Haruaki Nakaya; Tetsushi Furukawa

Female gender is a risk factor for drug‐induced arrhythmias associated with QT prolongation, which results mostly from blockade of the human ether‐a‐go‐go‐related gene (hERG) channel. Some clinical evidence suggests that oestrogen is a determinant of the gender‐differences in drug‐induced QT prolongation and baseline QTC intervals. Although the chronic effects of oestrogen have been studied, it remains unclear whether the gender differences are due entirely to transcriptional regulations through oestrogen receptors. We therefore investigated acute effects of the most bioactive oestrogen, 17β‐oestradiol (E2) at its physiological concentrations on cardiac repolarization and drug‐sensitivity of the hERG (IKr) channel in Langendorff‐perfused guinea pig hearts, patch‐clamped guinea pig cardiomyocytes and culture cells over‐expressing hERG. We found that physiological concentrations of E2 partially suppressed IKr in a receptor‐independent manner. E2‐induced modification of voltage‐dependence causes partial suppression of hERG currents. Mutagenesis studies showed that a common drug‐binding residue at the inner pore cavity was critical for the effects of E2 on the hERG channel. Furthermore, E2 enhanced both hERG suppression and QTC prolongation by its blocker, E4031. The lack of effects of testosterone at its physiological concentrations on both of hERG currents and E4031‐sensitivity of the hERG channel implicates the critical role of aromatic centroid present in E2 but not in testosterone. Our data indicate that E2 acutely affects the hERG channel gating and the E4031‐induced QTC prolongation, and may provide a novel mechanism for the higher susceptibility to drug‐induced arrhythmia in women.


The Journal of Neuroscience | 2007

Mode of Action and Functional Significance of Estrogen-Inducing Dendritic Growth, Spinogenesis, and Synaptogenesis in the Developing Purkinje Cell

Katsunori Sasahara; Hanako Shikimi; Shogo Haraguchi; Hirotaka Sakamoto; Shin-ichiro Honda; Nobuhiro Harada; Kazuyoshi Tsutsui

Neurosteroids are synthesized de novo from cholesterol in the brain. To understand neurosteroid action in the brain, data on the regio- and temporal-specific synthesis of neurosteroids are needed. Recently, we identified the Purkinje cell as an active neurosteroidogenic cell. In rodents, this neuron actively produces several neurosteroids including estradiol during neonatal life, when cerebellar neuronal circuit formation occurs. Estradiol may be involved in cerebellar neuronal circuit formation through promoting neuronal growth and neuronal synaptic contact, because the Purkinje cell expresses estrogen receptor-β (ERβ). To test this hypothesis, in this study we examined the effects of estradiol on dendritic growth, spinogenesis, and synaptogenesis in the Purkinje cell using neonatal wild-type (WT) mice or cytochrome P450 aromatase knock-out (ArKO) mice. Administration of estradiol to neonatal WT or ArKO mice increased dendritic growth, spinogenesis, and synaptogenesis in the Purkinje cell. In contrast, WT mice treated with tamoxifen, an ER antagonist, or ArKO mice exhibited decreased Purkinje dendritic growth, spinogenesis, and synaptogenesis at the same neonatal period. To elucidate the mode of action of estradiol, we further examined the expression of brain-derived neurotrophic factor (BDNF) in response to estrogen actions in the neonate. Estrogen administration to neonatal WT or ArKO mice increased the BDNF level in the cerebellum, whereas tamoxifen decreased the BDNF level in WT mice similar to ArKO mice. BDNF administration to tamoxifen-treated WT mice increased Purkinje dendritic growth. These results indicate that estradiol induces dendritic growth, spinogenesis, and synaptogenesis in the developing Purkinje cell via BDNF action during neonatal life.


Neuroendocrinology | 2003

Alteration in Sex-Specific Behaviors in Male Mice Lacking the Aromatase Gene

Takahiro Matsumoto; Shin-ichiro Honda; Nobuhiro Harada

Brain aromatase (P450arom) is a key enzyme in estrogen biosynthesis from testicular androgens. This local aromatization in neural tissues is thought to be an important process for sexual differentiation and activation of sexual behavior in male rodents. To determine the functional significance of the aromatase gene in development and activation of sex-specific behavior, we analyzed a series of behavioral profiles in gonadally intact male mice with targeted disruption of exons 1 and 2 of the aromatase gene (ArKO). In most cases, ArKO males were infertile and showed deficits in male sexual behavior including mount, intromission and ejaculation. Noncontact penile erection was not significantly affected by deletion of the aromatase gene. A great reduction of aggressive behavior against male intruders was also observed in ArKO males, while they tended to exhibit aggression toward estrous females during male copulatory tests. Furthermore, 73% of ArKO males showed infanticide toward pups, whereas characteristic parental behavior, but not infanticide, was observed in wild-type males. These results support the brain aromatization hypothesis and indicate that aromatase gene expression is a critical step not only for motivational and consummatory aspects of male sexual behavior, but also for aggressive and parental behaviors in male mice.


Molecular and Cellular Biochemistry | 2010

Fasting promotes the expression of SIRT1, an NAD+-dependent protein deacetylase, via activation of PPARα in mice

Satoru Hayashida; Akie Arimoto; Yukako Kuramoto; Tomohiro Kozako; Shin-ichiro Honda; Hiroshi Shimeno; Shinji Soeda

Calorie restriction (CR) extends lifespans in a wide variety of species. CR induces an increase in the NAD+/NADH ratio in cells and results in activation of SIRT1, an NAD+-dependent protein deacetylase that is thought to be a metabolic master switch linked to the modulation of lifespans. CR also affects the expression of peroxisome proliferator-activated receptors (PPARs). The three subtypes, PPARα, PPARγ, and PPARβ/δ, are expressed in multiple organs. They regulate different physiological functions such as energy metabolism, insulin action and inflammation, and apparently act as important regulators of longevity and aging. SIRT1 has been reported to repress the PPARγ by docking with its co-factors and to promote fat mobilization. However, the correlation between SIRT1 and other PPARs is not fully understood. CR initially induces a fasting-like response. In this study, we investigated how SIRT1 and PPARα correlate in the fasting-induced anti-aging pathways. A 24-h fasting in mice increased mRNA and protein expression of both SIRT1 and PPARα in the livers, where the NAD+ levels increased with increasing nicotinamide phosphoribosyltransferase (NAMPT) activity in the NAD+ salvage pathway. Treatment of Hepa1-6 cells in a low glucose medium conditions with NAD+ or NADH showed that the mRNA expression of both SIRT1 and PPARα can be enhanced by addition of NAD+, and decreased by increasing NADH levels. The cell experiments using SIRT1 antagonists and a PPARα agonist suggested that PPARα is a key molecule located upstream from SIRT1, and has a role in regulating SIRT1 gene expression in fasting-induced anti-aging pathways.


Journal of Biological Chemistry | 1999

A GCM Motif Protein Is Involved in Placenta-specific Expression of Human Aromatase Gene

Kazuyo Yamada; Hisamitsu Ogawa; Shin-ichiro Honda; Nobuhiro Harada; Tsuneko Okazaki

A new cis-element, trophoblast-specific element 2 (TSE2) is located in the placenta-specific enhancer of the human aromatase gene that dictates its tissue-specific expression. In the minimum enhancer region, an element similar to the trophoblast-specific element (TSE), originally described for the human chorionic gonadotropin α-subunit gene, also exists (Yamada, K., Harada, N., Honda, S., and Takagi, Y. (1995)J. Biol. Chem. 270, 25064–25069). The co-presence of TSE and TSE2 is required to direct trophoblast-specific expression driven by a heterologous thymidine kinase promoter. A 2562-base pair cDNA clone encoding a 436-amino acid protein that binds to TSE2 was isolated from a human placental cDNA library using a yeast one-hybrid system with the TSE2 as a reporter sequence. The protein was revealed to be identical to hGCMa, a mammalian homologue of theDrosophila GCM (glia cells missing) protein. Expression of hGCMa is restricted to the placenta. The protein also binds to PLE1 in the leptin promoter among other cis-elements reported to confer placenta-specific expression, suggesting that hGCMa is a placenta-specific transcription regulator, possibly involved in the expression of multiple placenta-specific genes.


Journal of Neuroendocrinology | 2002

Changes in the Arginine‐Vasopressin Immunoreactive Systems in Male Mice Lacking a Functional Aromatase Gene

L. Plumari; C. Viglietti-Panzica; F. Allieri; Shin-ichiro Honda; Nobuhiro Harada; Philippe Absil; Jacques Balthazart; Giancarlo Panzica

In male rodents, the arginine‐vasopressin‐immunoreactive (AVP‐ir) neurones of the bed nucleus of the stria terminalis (BNST) and medial amygdala are controlled by plasma testosterone levels (decreased after castration and restored by exogenous testosterone). AVP transcription in these nuclei is increased in adulthood by a synergistic action of the androgenic and oestrogenic metabolites of testosterone and, accordingly, androgen and oestrogen receptors are present in both BNST and medial amygdala. We used knockout mice lacking a functional aromatase enzyme (ArKO) to investigate the effects of a chronic depletion of oestrogens on the sexually dimorphic AVP system. Wild‐type (WT) and ArKO male mice were perfused 48 h after an i.c.v. colchicine injection and brain sections were then processed for AVP immunocytochemistry. A prominent decrease (but not a complete suppression) of AVP‐ir structures was observed in the BNST and medial amygdala of ArKO mice by comparison with the WT. Similarly, AVP‐ir fibres were reduced in the lateral septum of ArKO mice and but not in the medial preoptic area, a region where the AVP system is not sexually dimorphic in rats. No change was detected in the supraoptic and suprachiasmatic nuclei. However, a decrease in AVP‐ir cell numbers was however, detected in one subregion of the paraventricular nucleus. These data support the hypothesis that the steroid‐sensitive sexually dimorphic AVP system of the mouse forebrain is mainly under the control of aromatized metabolites of testosterone.

Collaboration


Dive into the Shin-ichiro Honda's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge