Network


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

Hotspot


Dive into the research topics where Hideyuki Mukai is active.

Publication


Featured researches published by Hideyuki Mukai.


Neuroscience Letters | 2010

Accumulation of tumor-suppressor PTEN in Alzheimer neurofibrillary tangles

Yuma Sonoda; Hideyuki Mukai; Kazuhiko Matsuo; Mikiko Takahashi; Yoshitaka Ono; Kiyoshi Maeda; Haruhiko Akiyama; Toshio Kawamata

The phosphatase and tensin homologue deleted on chromosome 10 (PTEN) negatively regulates intracellular levels of PIP3 and antagonizes the PI3K signaling pathway important for cell survival. The present study determined whether altered distribution of PTEN occurs in Alzheimers disease (AD) brains. We investigated a possible role for PTEN in postmortem brain tissues from elderly controls and patients with AD using immunoblotting and microscopic analyses. Intense immunolabeling was found in the large neurons such as pyramidal cells. In normal neurons, PTEN was located in the nucleus, the cytoplasm of cell bodies and the proximal portion of apical dendrites. Reduced expression and redistribution of PTEN was seen in the remaining neurons in AD. In addition, PTEN was redistributed in damaged neurons from the nucleus and cytoplasm to neuritic pathology such as intracellular neurofibrillary tangles (NFTs), neuropil threads and dystrophic neurites within senile plaques in AD hippocampus, subiculum, entorhinal cortex and angular gyrus. Furthermore, double immunofluorescence staining showed dual labeling of intracellular NFTs for PTEN and tau, labeling of some axons for PTEN and phosphorylated neurofilament, and weak labeling of a few reactive astrocytes around senile plaques for PTEN and GFAP. Double labeling of NFTs was observed in a subset of tangle-bearing neurons either for PTEN and GSK3beta or for PTEN and MEK. Thus our results suggest that PTEN delocalized from the nucleus to the cytoplasm and to intracellular NFTs may cause a deregulation of PI3K pathway in the cytoplasm and may induce the nuclear dysfunction of PTEN in AD degenerating neurons.


Genes to Cells | 2004

Centrosome-targeting region of CG-NAP causes centrosome amplification by recruiting cyclin E-cdk2 complex

Tamako Nishimura; Mikiko Takahashi; Hon-Song Kim; Hideyuki Mukai; Yoshitaka Ono

Centrosome duplication occurs once per cell cycle and is thought to be triggered by cyclin E‐cdk2. However, it is largely unknown how the duplication is regulated. Here, we found that the expression of the centrosome‐targeting region of CG‐NAP (centrosome and Golgi‐localized PKN‐associated protein), which we designate as CG‐NAP/D, increased the number of centrosomes in Chinese hamster ovary (CHO)‐K1 cells. The amplified centrosomes co‐localized with centrosome markers γ‐tubulin, centrin‐2 and kendrin as well as endogenous CG‐NAP. When CG‐NAP/D was dislocated from centrosomes by deleting the centrosome‐targeting domain or by fusing with a membrane‐targeting sequence, centrosome amplification was suppressed. CG‐NAP/D interacted with exogenously expressed cyclin E, which co‐localized at centrosomes. The immunoprecipitates of CG‐NAP/D exhibited histone H1 kinase activity, suggesting the co‐immunoprecipitation of active cyclin‐cdk complexes. Furthermore, centrosome fractions prepared from cells expressing CG‐NAP/D contained increased amount of cdk2 compared with those from control cells. Centrosome amplification by CG‐NAP/D was suppressed by co‐expression of a mutant cyclin E unable to interact with cdk2. These results suggest that CG‐NAP/D causes centrosome amplification by anchoring excess amount of cyclin E‐cdk2 to centrosomes and, possibly, CG‐NAP participates in centrosome duplication by recruiting cyclin E‐cdk2 to centrosomes in normal cell cycle.


Journal of Biological Chemistry | 2005

The role of the regulatory subunit of fission yeast calcineurin for in vivo activity and its relevance to FK506 sensitivity

Susie Ong Sio; Takafumi Suehiro; Reiko Sugiura; Mai Takeuchi; Hideyuki Mukai; Takayoshi Kuno

Calcineurin, a protein phosphatase required for Ca2+signaling in many cell types, is a heterodimer composed of catalytic and regulatory subunits. The fission yeast genome encodes a single set of catalytic (Ppb1) and regulatory (Cnb1) subunits, providing an ideal model system to study the functions of these subunits in vivo. Here, we cloned the cnb1+ gene and showed that the cnb1 knock-out (Δcnb1) exhibits identical phenotypes with Δppb1 and that overexpression of Ppb1 failed to suppress the phenotypes of Δcnb1. Interestingly, overexpression of the C-terminal-deleted Ppb1 (Ppb1ΔC), the constitutively active form of Ppb1, also failed to suppress the phenotypes of Δcnb1. FK506 caused MgCl2 sensitivity to the wild-type cells in an FKBP12-dependentmanner. Co-overexpression of Ppb1 and Cnb1 suppressed the FK506-induced MgCl2 sensitivity, but the suppression was only partial, suggesting that an excess amount of the Ppb1-Cnb1 complex cannot compete out the FKBP12-FK506 complex. Although overexpression of Ppb1ΔC alone had little effect on cell growth, co-overexpression of Ppb1ΔC and Cnb1 caused a distinct growth defect. FK506 suppressed the growth defect when Cnb1 was co-expressed using the attenuated nmt1 promoter, but it failed to suppress the defect when Cnb1 was co-expressed using the wild-type nmt1 promoter. Knock-out of the prz1+ gene, encoding a downstream target transcription factor of calcineurin, suppressed the growth defect irrespective of the promoter potency. These results suggest that Cnb1 is essential for the activation of calcineurin and that the activated calcineurin is the pharmacological target of the FKBP12-FK506 complex in vivo.


Neuroscience Letters | 2009

A peptidyl-prolyl isomerase, FKBP12, accumulates in Alzheimer neurofibrillary tangles.

Hisato Sugata; Kazuhiko Matsuo; Taisuke Nakagawa; Mikiko Takahashi; Hideyuki Mukai; Yoshitaka Ono; Kiyoshi Maeda; Haruhiko Akiyama; Toshio Kawamata

We investigated a possible role in Alzheimers disease (AD) for FKBP12, a peptidyl-prolyl cis-trans isomerase known to be important in protein assembly, folding and transportation by using Western blotting and microscopic analyses in postmortem brain tissues from elderly controls and the patients with AD. FKBP12 was enriched and localized to neuronal cell bodies and neurites in control brains. Intense immunoreactivity was found in large neurons such as pyramidal cells. Many FKBP12 positive granules were located in the cytoplasm and the proximal portion of dendrites and axons, and in the nuclei. By contrast, the expression of FKBP12 in AD brains was lower than in control brains. Furthermore, numerous intracellular neurofibrillary tangles (NFTs) were stained for FKBP12 in the hippocampal CA1 subfield, subiculum, entorhinal cortex and angular gyrus. Neuritic pathology such as neuropil threads and dystrophic neurites (DNs) within senile plaques (SPs) and some reactive astrocytes were also immunolabeled for FKBP12 in AD. Double immunofluorescence staining showed dual labeling of intracellular NFTs for FKBP12 and tau. Similar results were obtained in reactive astrocytes for the combination of FKBP12 and glial fibrillary acidic protein (GFAP). Labeling for FKBP12 was dense in axons stained for highly phosphorylated neurofilament protein. Thus our results suggest that FKBP12 may be involved in neuronal or astrocytic cytoskeletal organization and in the abnormal metabolism of tau protein in AD damaged neurons.


Oncogene | 2008

Calcineurin-mediated dephosphorylation of c-Jun Ser-243 is required for c-Jun protein stability and cell transformation

Chi Chen Huang; Ju Ming Wang; Ushio Kikkawa; Hideyuki Mukai; Meng Ru Shen; Ikuo Morita; Ben Kuen Chen; Wen-Chang Chang

The proto-oncogene c-Jun plays an important role in regulating tumor progression. We previously reported that the serine/threonine phosphatase calcineurin (CaN, also called PP2B) dephosphorylates the C-terminus (Ser-243) of c-Jun, resulting in the increase in c-Jun and Sp1 interaction, and subsequent c-Jun-induced gene expression. Here, we demonstrate the interaction of c-Jun and CaN in the nucleus of living cells by fluorescence resonance energy transfer assay and that this interaction is mediated through the calmodulin-binding domain of CaN. Furthermore, c-Jun protein stability was altered by CaN-mediated dephosphorylation at the Ser-243 site of c-Jun. The half-life of the c-Jun mutant, c-Jun-S243A was longer than that of the wild-type c-Jun. Moreover, silencing of endogenous CaN expression led to increased c-Jun ubiquitination and decreased stability. In 46% of clinical cervical tissue samples obtained from patients with cervical cancer, enhanced c-Jun and CaN expression, as well as decreased phospho-Ser-243 expression levels were detected. Our results suggest that CaN stabilizes c-Jun by dephosphorylating c-Jun at Ser-243 to enhance its tumorigenic ability.


American Journal of Physiology-heart and Circulatory Physiology | 2011

Hypotonic swelling-induced activation of PKN1 mediates cell survival in cardiac myocytes

Katsuya Kajimoto; Dan Shao; Hiromitsu Takagi; Gregorio Maceri; Daniela Zablocki; Hideyuki Mukai; Yoshitaka Ono; Junichi Sadoshima

Hypotonic cell swelling in the myocardium is induced by pathological conditions, including ischemia-reperfusion, and affects the activities of ion transporters/channels and gene expression. However, the signaling mechanism activated by hypotonic stress (HS) is not fully understood in cardiac myocytes. A specialized protein kinase cascade, consisting of Pkc1 and MAPKs, is activated by HS in yeast. Here, we demonstrate that protein kinase N1 (PKN1), a serine/threonine protein kinase and a homolog of Pkc1, is activated by HS (67% osmolarity) within 5 min and reaches peak activity at 60 min in cardiac myocytes. Activation of PKN1 by HS was accompanied by Thr(774) phosphorylation and concomitant activation of PDK1, a potential upstream regulator of PKN1. HS also activated RhoA, thereby increasing interactions between PKN1 and RhoA. PP1 (10(-5) M), a selective Src family tyrosine kinase inhibitor, significantly suppressed HS-induced activation of RhoA and PKN1. Constitutively active PKN1 significantly increased the transcriptional activity of Elk1-GAL4, an effect that was inhibited by dominant negative MEK. Overexpression of PKN1 significantly increased ERK phosphorylation, whereas downregulation of PKN1 inhibited HS-induced ERK phosphorylation. Downregulation of PKN1 and inhibition of ERK by U-0126 both significantly inhibited the survival of cardiac myocytes in the presence of HS. These results suggest that a signaling cascade, consisting of Src, RhoA, PKN1, and ERK, is activated by HS, thereby promoting cardiac myocyte survival.


Biochemical Journal | 2010

Development of an intracellularly acting inhibitory peptide selective for PKN

Kazuhiro Shiga; Kentaro Takayama; Shiroh Futaki; Jessica E. Hutti; Lewis C. Cantley; Katsuko Ueki; Yoshitaka Ono; Hideyuki Mukai

PKNs form a subfamily of the AGC serine/threonine protein kinases, and have a catalytic domain homologous with that of PKC (protein kinase C) in the C-terminal region and three characteristic ACC (antiparallel coiled-coil) domain repeats in the N-terminal region. The preferred peptide phosphorylation motif for PKNs determined by a combinatorial peptide library method was highly similar to that of PKCs within a 10-amino-acid stretch. Previously reported PKN inhibitory compounds also inhibit PKCs to a similar extent, and no PKN selective inhibitors have been commercially available. We have identified a 15-amino-acid peptide inhibitor of PKNs based on amino acids 485-499 of the C-terminal region of the C2-like domain of PKN1. This peptide, designated as PRL, selectively inhibits the kinase activity of all isoforms of PKN (Ki=0.7 muM) towards a peptide substrate, as well as autophosphorylation activity of PKN in vitro, in contrast with PKC. Reversible conjugation by a disulfide bond of a carrier peptide bearing a penetration accelerating sequence to PRL, facilitated the cellular uptake of this peptide and significantly inhibited phosphorylation of tau by PKN1 at the PKN1-specific phosphorylation site in vivo. This peptide may serve as a valuable tool for investigating PKN activation and PKN-mediated responses.


Molecular Carcinogenesis | 2005

Involvement of protein kinase PKN1 in G2/M delay caused by arsenite

Takayuki Isagawa; Mikiko Takahashi; Tomohisa Kato; Hideyuki Mukai; Yoshitaka Ono

PKN1 is a serine/threonine protein kinase that has been reported to mediate cellular response to stress. We show here that in response to arsenite exposure, PKN1 kinase activity was stimulated, which was associated with increased binding of PKN1 to Cdc25C and delayed mitotic entry. A role for PKN1 in mediating arsenite‐induced G2/M delay was supported by the finding that expression of a constitutively active form of PKN1 (PKN1AF3) in HeLa cells delayed the mitotic entry of cell cycle. Further experiments indicate that PKN1 directly phosphorylated serine 216 (Ser216) in Cdc25C, which then facilitated association between Cdc25C and 14‐3‐3. Significantly, expression of a phosphorylation mutant of Cdc25C (S216A) partially abrogated the cell‐cycle arrest in response to arsenite. Together, our results suggest that PKN1 mediates arsenite‐induced delay of the G2/M transition by binding to and phoshorylating Cdc25C.


Scientific Reports | 2016

PKN3 is the major regulator of angiogenesis and tumor metastasis in mice

Hideyuki Mukai; Aiko Muramatsu; Rana Mashud; Koji Kubouchi; Sho Tsujimoto; Tsunaki Hongu; Yasunori Kanaho; Masanobu Tsubaki; Shozo Nishida; Go Shioi; Sally Danno; Mona Mehruba; Ryosuke Satoh; Reiko Sugiura

PKN, a conserved family member related to PKC, was the first protein kinase identified as a target of the small GTPase Rho. PKN is involved in various functions including cytoskeletal arrangement and cell adhesion. Furthermore, the enrichment of PKN3 mRNA in some cancer cell lines as well as its requirement in malignant prostate cell growth suggested its involvement in oncogenesis. Despite intensive research efforts, physiological as well as pathological roles of PKN3 in vivo remain elusive. Here, we generated mice with a targeted deletion of PKN3. The PKN3 knockout (KO) mice are viable and develop normally. However, the absence of PKN3 had an impact on angiogenesis as evidenced by marked suppressions of micro-vessel sprouting in ex vivo aortic ring assay and in vivo corneal pocket assay. Furthermore, the PKN3 KO mice exhibited an impaired lung metastasis of melanoma cells when administered from the tail vein. Importantly, PKN3 knock-down by small interfering RNA (siRNA) induced a glycosylation defect of cell-surface glycoproteins, including ICAM-1, integrin β1 and integrin α5 in HUVECs. Our data provide the first in vivo genetic demonstration that PKN3 plays critical roles in angiogenesis and tumor metastasis, and that defective maturation of cell surface glycoproteins might underlie these phenotypes.


Methods in Enzymology | 2006

Purification and Kinase Assay of PKN

Hideyuki Mukai; Yoshitaka Ono

PKN is a serine/threonine protein kinase, which has a catalytic domain highly homologous to that of protein kinase C (PKC) in the carboxyl-terminal region and three repeats of the antiparallel coiled coil (ACC) domain in the amino-terminal region. Mammalian PKN has three isoforms each derived from different genes, PKN1 (PKNalpha/PRK1/PAK1), PKN2 (PRK2/PAK2/PKNgamma), and PKN3 (PKNbeta). PKN isoforms show different enzymatic properties and tissue distributions and have been implicated in various distinct cellular processes (reviewed in Mukai [2003]). This chapter discusses methods to prepare purified enzymes and to assay substrate phosphorylation activities.

Collaboration


Dive into the Hideyuki Mukai's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Haruhiko Akiyama

Institute of Medical Science

View shared research outputs
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