Network


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

Hotspot


Dive into the research topics where Megumi Hirose is active.

Publication


Featured researches published by Megumi Hirose.


Proceedings of the National Academy of Sciences of the United States of America | 2001

Altered psychomotor behaviors in mice lacking pituitary adenylate cyclase-activating polypeptide (PACAP)

Hitoshi Hashimoto; Norihito Shintani; Kazuhiro Tanaka; Wakaba Mori; Megumi Hirose; Toshio Matsuda; Masaki Sakaue; Jun-ichi Miyazaki; Hitoshi Niwa; Fumi Tashiro; Kyohei Yamamoto; Kazumi Koga; Shuhei Tomimoto; Akiyoshi Kunugi; Sayoko Suetake; Akemichi Baba

Pituitary adenylate cyclase-activating polypeptide (PACAP) has been conserved remarkably during evolution and is widely expressed in the mammalian brain. In Drosophila, mutation of the PACAP homologue results in behavioral defects, including impaired olfaction-associated learning and changes in ethanol sensitivity. Here, we report the generation of mice lacking the PACAP gene (PACAP−/−). PACAP−/− mice were born in the expected Mendelian ratios but had a high early-mortality rate. The surviving adult PACAP−/− mice displayed remarkable behavioral changes; they exhibited hyperactive and explosive jumping behaviors in an open field, increased exploratory behavior, and less anxiety in the elevated plus maze, emergence, and novel-object tests. Analysis of PACAP−/− mice brains revealed that the serotonin metabolite 5-hydroxyindoleacetic acid was slightly decreased in the cortex and striatum compared with wild-type mice. The present study provides evidence that PACAP plays a previously uncharacterized role in the regulation of psychomotor behaviors.


Regulatory Peptides | 2002

Defects in reproductive functions in PACAP-deficient female mice.

Norihito Shintani; Wakaba Mori; Hitoshi Hashimoto; Masayuki Imai; Kazuhiro Tanaka; Shuhei Tomimoto; Megumi Hirose; Chihiro Kawaguchi; Akemichi Baba

Pituitary adenylate cyclase-activating polypeptide (PACAP) is a highly conserved neuropeptide and widely expressed in both brain and peripheral tissues, including several reproductive organs (e.g., testis and ovary). PACAP stimulates syntheses of several sexual hormones and steroids, suggesting it has possible roles in reproductive function. In this study, the role of PACAP in female reproductive functions such as fertility, mating behavior and maternal behaviors were investigated by using mice lacking PACAP (PACAP(-/-)). PACAP(-/-) females showed reduced fertility (the number of parturitions relative to the number of pairings). Mating experiments using vasectomized males revealed that mating frequency and its intervals in some PACAP(-/-) females were quite different (zero to eight times/4 weeks), whereas the frequency was relatively constant (two to three times/4 weeks) in wild-type females. In PACAP(-/-) females, maternal crouching behavior tended to decrease compared to wild-type females, although the influence of litter size on maternal behavior needs to be considered. These data suggest a role for endogenous PACAP in female reproductive processes.


Regulatory Peptides | 2005

Neuroprotective action of endogenous PACAP in cultured rat cortical neurons

Norihito Shintani; Sayoko Suetake; Hitoshi Hashimoto; Kazumi Koga; Atsushi Kasai; Chihiro Kawaguchi; Yoshiko Morita; Megumi Hirose; Yoshiyuki Sakai; Shuhei Tomimoto; Toshio Matsuda; Akemichi Baba

Pituitary adenylate cyclase-activating polypeptide (PACAP) exerts neurotrophic effects both in vitro and in vivo. Here we demonstrate the upregulation of PACAP mRNA expression in cultured rat cortical neurons after excitotoxic glutamate exposure, and the exacerbating effect of the PACAP receptor antagonist, PACAP(6-38), on neuronal viability. PACAP mRNA levels were increased up to 3.5-fold 8 h after glutamate exposure. PACAP(6-38) decreased the viability of cortical neurons, irrespective of whether the cells were exposed to glutamate or not. PACAP(6-38) also inhibited glutamate-induced expression of PACAP mRNA, suggesting that PACAP acts via an autocrine or paracrine mechanism to enhance PACAP expression itself. Glutamate exposure is known to increase brain-derived neurotrophic factor (BDNF) mRNA expression. This increased expression was markedly suppressed by PACAP(6-38). Our previous study has shown that PACAP stimulates the PACAP gene transcription in PC12 cells. Taken together, these data may suggest that endogenous PACAP regulates the expression of PACAP itself and BDNF. Although it may also be possible that PACAP(6-38)-induced death of PACAP and BDNF mRNA-expressing cells, per se, results in reduced levels of these mRNAs, the present results support the idea that endogenous PACAP has a neuroprotective action.


Reproductive Biomedicine Online | 2008

Stable embryonic stem cell lines in rabbits: potential small animal models for human research

Arata Honda; Michiko Hirose; Kimiko Inoue; Narumi Ogonuki; Hiromi Miki; Nobuhiro Shimozawa; Masanori Hatori; Natsumi Shimizu; Takehide Murata; Megumi Hirose; Kazufumi Katayama; Noriko Wakisaka; Hiroyuki Miyoshi; Kazunari K. Yokoyama; Tadashi Sankai; Atsuo Ogura

Although embryonic stem (ES) cell lines derived from mice and primates are used extensively, the development of such lines from other mammals is extremely difficult because of their rapid decline in proliferation potential and pluripotency after several passages. This study describes the establishment of rabbit ES cell lines with indefinite proliferation potential. It was found that the feeder cell density determines the fate of rabbit ES cells, and that maximum proliferation potential was obtained when they were cultured on a feeder cell density of one-sixth of the density at confluency. Higher and lower densities of feeder cells induced ES cell differentiation or division arrest. Under optimized conditions, rabbit ES cells were passaged 50 times, after which they still possessed high telomerase activity. This culture system enabled efficient gene transduction and clonal expansion from single cells. During culture, rabbit ES cells exhibited flattened monolayer cell colonies, as reported for monkey and human ES cells, and expressed pluripotency markers. Embryoid bodies and teratomas formed readily in vitro and in vivo respectively. These ES cell lines can be safely cryopreserved for later use. Thus, rabbit ES cells can be added to the list of stable mammalian ES cells, enabling the rabbit to be used as a small animal model for the study of human cell transplantation therapy.


Regulatory Peptides | 2002

Involvement of intracellular Ca2+ elevation but not cyclic AMP in PACAP-induced p38 MAP kinase activation in PC12 cells.

Yoshiyuki Sakai; Hitoshi Hashimoto; Norihito Shintani; Ayako Ichibori; Shuhei Tomimoto; Kazuhiro Tanaka; Megumi Hirose; Akemichi Baba

We have recently shown that in PC12 cells, pituitary adenylate cyclase-activating polypeptide (PACAP) and NGF synergistically stimulate PACAP mRNA expression primarily via a mechanism involving a p38 mitogen-activated protein kinase (MAPK)-dependent pathway. Here we have analyzed p38 MAPK activation by PACAP and the mechanism underlying this action of PACAP in PC12 cells. PACAP increased phosphorylation of p38 MAPK with a bell-shaped dose-response relationship and a maximal effect was obtained at 10(-8) M. PACAP (10(-8) M)-induced p38 MAPK phosphorylation was already evident at 2.5 min, maximal at 5 min, and rapidly declined thereafter. PACAP-induced p38 MAPK phosphorylation was potently inhibited by depletion of Ca(2+) stores with thapsigargin and partially inhibited by the phospholipase C inhibitor U-73122, L-type voltage-dependent calcium channel inhibitors nifedipine and nimodipine, and the Ca(2+) chelator EGTA, whereas the protein kinase C inhibitor calphostin C, the protein kinase A inhibitor H-89, the cAMP antagonist Rp-cAMP, and the nonselective cation channel blocker SKF96365 had no effect. These results indicate that PACAP activates p38 MAPK in PC12 cells through activation of a phospholipase C, mobilization of intracellular Ca(2+) stores, and Ca(2+) influx through voltage-dependent Ca(2+) channels, but not cyclic AMP-dependent mechanisms.


Regulatory Peptides | 2005

Differential expression of mRNAs for PACAP and its receptors during neural differentiation of embryonic stem cells.

Megumi Hirose; Hitoshi Hashimoto; Norihito Shintani; Megumi Nakanishi; Naohisa Arakawa; Junko Iga; Hitoshi Niwa; Jun-ichi Miyazaki; Akemichi Baba

The expressions of mRNAs for pituitary adenylate cyclase-activating polypeptide (PACAP), vasoactive intestinal peptide (VIP), and their receptors (PAC1, VPAC1 and VPAC2) were examined in the five steps of the in vitro neuronal culture model of embryonic stem (ES) cell differentiation. mRNAs for PACAP, VIP, PAC1 receptor, and VPAC2 receptor were moderately expressed in neural stem cell-enriched cultures, while VPAC1 receptor mRNA was most prominently expressed in embryoid bodies (EBs). The expression of PAC1 receptor mRNA was further upregulated after terminal differentiation into neurons. In contrast, the expressions of PAC1 receptor and PACAP mRNAs were markedly decreased after glial differentiation. These results suggest that this in vitro neuronal culture system will be a useful model for future studies on the functional role of the PACAPergic system during different stages of neuronal development.


Annals of the New York Academy of Sciences | 2006

Inhibition of Self‐Renewal and Induction of Neural Differentiation by PACAP in Neural Progenitor Cells

Megumi Hirose; Hitoshi Hashimoto; Junko Iga; Norihito Shintani; Megumi Nakanishi; Naohisa Arakawa; Takeshi Shimada; Akemichi Baba

Abstract:  Several lines of evidence have suggested roles for pituitary adenylate cyclase‐activating polypeptide (PACAP) in the developing nervous system. Previously, we showed that mRNA for PACAP, vasoactive intestinal peptide (VIP), and their three receptor subtypes, is differentially expressed in embryonic stem (ES) cells, ES cell‐derived, neural stem cell‐enriched cultures, and differentiated neurons, by using the five steps of the in vitro neuronal culture model of ES cell differentiation . Here, we examined the effects of PACAP on self‐renewal and cell lineage determination of neural progenitor/stem cells. PACAP inhibited the basic fibroblast growth factor‐induced proliferation (self‐renewal), as assessed by neurosphere formation. PACAP increased microtubule‐associated protein 2‐positive neurons without affecting the number of cells positive for the neural stem cell marker nestin, astrocyte marker glial fibrillary acidic protein, and oligodendrocyte marker CNPase. These results suggest that PACAP inhibits self‐renewal but, instead, induces early neuronal differentiation of neural progenitor cells.


Journal of Gene Medicine | 2005

A database of recombinant viruses and recombinant viral vectors available from the RIKEN DNA bank

Hideyo Ugai; Takehide Murata; Yoshinori Nagamura; Yoshihiro Ugawa; Erika Suzuki; Hatsumi Nakata; Yukari Kujime; Sanae Inamoto; Megumi Hirose; Kumiko Inabe; Miho Terashima; Takahito Yamasaki; Bingbing Liu; Koji Nakade; Jianzhi Pan; Makoto Kimura; Izumu Saito; Hirofumi Hamada; Yuichi Obata; Kazunari K. Yokoyama

Viral vectors are required as gene‐delivery systems for gene therapy and basic research. Recombinant adenoviruses (rAds) expressing genes of interest are being developed as research tools and many studies in vitro and in vivo have already been performed with such rAds.


Biochemical and Biophysical Research Communications | 2001

Involvement of p38 MAP kinase pathway in the synergistic activation of PACAP mRNA expression by NGF and PACAP in PC12h cells.

Yoshiyuki Sakai; Hitoshi Hashimoto; Norihito Shintani; Shuhei Tomimoto; Kazuhiro Tanaka; Ayako Ichibori; Megumi Hirose; Akemichi Baba


Biochemical and Biophysical Research Communications | 2005

Purification of infectious adenovirus in two hours by ultracentrifugation and tangential flow filtration

Hideyo Ugai; Takahito Yamasaki; Megumi Hirose; Kumiko Inabe; Yukari Kujime; Miho Terashima; Bingbing Liu; Hong Tang; Mujun Zhao; Takehide Murata; Makoto Kimura; Jianzhi Pan; Yuichi Obata; Hirofumi Hamada; Kazunari K. Yokoyama

Collaboration


Dive into the Megumi Hirose's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Takehide Murata

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hideyo Ugai

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge