Network


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

Hotspot


Dive into the research topics where Noriyuki Murai is active.

Publication


Featured researches published by Noriyuki Murai.


Journal of Biological Chemistry | 1996

GroEL Locked in a Closed Conformation by an Interdomain Cross-link Can Bind ATP and Polypeptide but Cannot Process Further Reaction Steps

Noriyuki Murai; Yoshihide Makino; Masasuke Yoshida

It has been believed that when GroEL binds to GroES its apical domain moves upward and outward. To inhibit this “opening” movement, its equatorial and apical domains were cross-linked through a disulfide bond between mutationally introduced cysteine residues at the positions of Asp-83 and Lys-327. To avoid possible undesired cross-linking, we at first prepared a mutant GroEL (GroELNC; Cys-138 → Ser, Cys-458 → Ser, Cys-519 → Ser) in which all cysteine residues in wild-type GroEL were replaced by serine residues. GroELNC was fully functional as a chaperonin. We then introduced the above two point mutations into GroELNC to generate a mutant (GroELAEX; Cys-138 → Ser, Cys-458 → Ser, Cys-519 → Ser and Asp-83 → Cys, Lys-327 → Cys). Oxidized GroELAEX, which is locked in a “closed” conformation by an interdomain disulfide bond, can bind 6-7 mol of ATP, which remain bound without hydrolysis. This ATP-bound, oxidized GroELAEX can bind the stably nonnative substrate protein isopropylmalate dehydrogenase, whereas the nucleotide-free oxidized GroELAEX binds it with a weaker affinity. However, oxidized GroELAEX fails to process further reaction steps such as ATP hydrolysis, binding of GroES, dissociation of substrate protein from GroEL, and facilitating protein folding. When disulfide bonds in oxidized GroELAEX are reduced, GroELAEX exerts the ability to process all the reactions just as GroELNC and wild-type GroEL. Indications from these results are: hydrolysis of ATP may require opening movement of the apical domain; GroES binds to an open form of GroEL; and substrate polypeptide is released from GroEL coupled with either ATP hydrolysis or opening movement of the apical domain.


Journal of Biological Chemistry | 2003

Identification of Nuclear Export Signals in Antizyme-1

Noriyuki Murai; Yasuko Murakami; Senya Matsufuji

Antizyme-1 (AZ1) is a protein that negatively regulates polyamine synthesis by inhibiting the key synthetic enzyme ornithine decarboxylase and targeting it for degradation by the 26 S proteasome. Recent work shows that antizyme protein translocates to the nucleus during mouse development (Gritli-Linde, A., Nilssom, J., Bohlooly, Y. M., Heby, O., and Linde, A. (2001) Dev. Dyn. 220, 259-275). However, the significance and mechanism of this phenomenon remain unclear. In this study, we expressed AZ1 fused with enhanced green fluorescent protein (EGFP) to study its localization in a living cell. We found that EGFP-AZ1 was predominantly localized in the cytoplasm and that treatment with leptomycin B, a specific inhibitor of chromosomal region maintenance 1 (CRM1) induced nuclear accumulation of EGFP-AZ1 in Chinese hamster ovary and NIH3T3 cells. Two independent nuclear export signal (NES) sequences, each containing essential hydrophobic residues, were identified in the 50 N-terminal amino acid residues and in the central part of AZ1. The activity of the second NES was inhibited by an N-terminal adjacent region and was only revealed in N-terminal truncated constructs. Both NESs were active when fused to an artificial nuclear protein SV40-NLS-EGFP-EGFP. The ability of AZ1 to shuttle between the nucleus and the cytoplasm suggests that it has a novel function in the nucleus.


Kidney International | 2013

Rho-kinase inhibition prevents the progression of diabetic nephropathy by downregulating hypoxia-inducible factor 1α

Keiichiro Matoba; Daiji Kawanami; Rina Okada; Masami Tsukamoto; Jun Kinoshita; Tomoko Ito; Sho Ishizawa; Yasushi Kanazawa; Tamotsu Yokota; Noriyuki Murai; Senya Matsufuji; Junko Takahashi-Fujigasaki; Kazunori Utsunomiya

The small GTPase Rho and its effector Rho-kinase are involved in the pathogenesis of diabetic nephropathy. Accumulating evidence shows that hypoxia-inducible factor-1α (HIF-1α) is a key regulator of renal sclerosis under diabetic conditions. However, the interactions of Rho-kinase and HIF-1α in the development of renal dysfunction have not been defined. Here, we assessed whether Rho-kinase blockade attenuates HIF-1α induction and the subsequent fibrotic response using type 2 diabetic mice and cultured mesangial cells. Fasudil, a Rho-kinase inhibitor, reduced urinary albumin excretion, mesangial matrix expansion, and the expression of fibrotic mediators in db/db mice. Mechanistically, HIF-1α accumulation and the expression of its target genes that contribute to diabetic glomerulosclerosis were also prevented by fasudil in the renal cortex. In mesangial cells, Rho/Rho-kinase signaling was activated under hypoxic conditions. Further in vitro studies showed that pharmacological and genetic inhibition of Rho-kinase promoted proteasomal HIF-1α degradation, which subsequently suppressed HIF-1-dependent profibrotic gene expression by upregulation of prolyl hydroxylase 2. Thus, we found a previously unrecognized renoprotective mechanism for the effects of Rho-kinase inhibition and this could be a potential therapeutic target for the treatment of diabetic nephropathy.


Journal of Biological Chemistry | 1995

KINETIC ANALYSIS OF INTERACTIONS BETWEEN GROEL AND REDUCED ALPHA -LACTALBUMIN : EFFECT OF GROES AND NUCLEOTIDES

Noriyuki Murai; Hideki Taguchi; Masasuke Yoshida

The real-time analysis of the association and dissociation of chaperonin with respect to its substrate protein was carried out using the BIAcore


Biochimica et Biophysica Acta | 2002

Regulation of ornithine decarboxylase by antizymes and antizyme inhibitor in zebrafish (Danio rerio)

Tomasz Hascilowicz; Noriyuki Murai; Senya Matsufuji; Yasuko Murakami

Mammalian polyamine synthesis is regulated by a unique feedback mechanism. When cellular polyamine levels increase, antizyme, an ornithine decarboxylase (ODC) inhibitory protein, is induced by polyamine-dependent translational frameshifting. Antizyme not only inhibits ODC, a key enzyme in polyamine synthesis, it also targets the enzyme degradation by the 26S proteasome. Furthermore, it suppresses cellular uptake of polyamines. Previously, we isolated two zebrafish antizymes with different expressions and activities. This suggested that a common feedback mechanism of polyamine metabolism might operate in mammals and zebrafish (Danio rerio). In the present study, cDNAs of zebrafish ODC and antizyme inhibitor, another regulatory protein that inhibits antizyme action, were cloned. The presence of ODC and antizyme inhibitor mRNAs was confirmed by Northern blotting in embryos and adult fish, as well as in a zebrafish-derived cell line (BRF41). The activity of the ODC cDNA expression product was inhibited by short and long zebrafish antizymes, and recombinant zebrafish antizyme inhibitor reversed this inhibition. In the BRF41 cells, the ODC half-life was considerably longer than that of mammalian ODC but shorter than that of Schizosaccharomyces pombe. Spermidine elicited a rapid decay of ODC activity and ODC protein in a protein synthesis-dependent manner.


Experimental Cell Research | 2009

The change of antizyme inhibitor expression and its possible role during mammalian cell cycle

Yasuko Murakami; Jun-ichiro Suzuki; Keijiro Samejima; Kenjiro Kikuchi; Tomasz Hascilowicz; Noriyuki Murai; Senya Matsufuji; Takami Oka

Antizyme inhibitor (AIn), a homolog of ODC, binds to antizyme and inactivates it. We report here that AIn increased at the G1 phase of the cell cycle, preceding the peak of ODC activity in HTC cells in culture. During interphase AIn was present mainly in the cytoplasm and turned over rapidly with the half-life of 10 to 20 min, while antizyme was localized in the nucleus. The level of AIn increased again at the G2/M phase along with ODC, and the rate of turn-over of AIn in mitotic cells decreased with the half-life of approximately 40 min. AIn was colocalized with antizyme at centrosomes during the period from prophase through late anaphase and at the midzone/midbody during telophase. Thereafter, AIn and antizyme were separated and present at different regions on the midbody at late telophase. AIn disappeared at late cytokinesis, whereas antizyme remained at the cytokinesis remnant. Reduction of AIn by RNA interference caused the increase in the number of binucleated cells in HTC cells in culture. These findings suggested that AIn contributed to a rapid increase in ODC at the G1 phase and also played a role in facilitating cells to complete mitosis during the cell cycle.


Journal of Cellular Biochemistry | 2009

Subcellular Localization and Phosphorylation of Antizyme 2

Noriyuki Murai; Akihiro Shimizu; Yasuko Murakami; Senya Matsufuji

Antizymes (AZs) are polyamine‐induced proteins that negatively regulate cellular polyamine synthesis and uptake. Three antizyme isoforms are conserved among mammals. AZ1 and AZ2 have a broad tissue distribution, while AZ3 is testis specific. Both AZ1 and AZ2 inhibit ornithine decarboxylase (ODC) activity by binding to ODC monomer and target it to the 26S proteasome at least in vivo. Both also inhibit extra‐cellular polyamine uptake. Despite their being indistinguishable by these criteria, we show here using enhanced green fluorescent protein (EGFP)‐AZ2 fusion protein that in mammalian cells, the subcellular location of AZ2 is mainly in the nucleus, and is different from that of AZ1. The C‐terminal part of AZ2 is necessary for the nuclear distribution. Within a few hours, a shift in the distribution of EGFP‐AZ2 fusion protein from cytoplasm to the nucleus or from nucleus to cytoplasm is observable in NIH3T3 cells. In addition, we found that in cells a majority of AZ2, but not AZ1, is phosphorylated at Ser‐186, likely by protein kinase CK2. There may be a specific function of AZ2 in the nucleus. J. Cell. Biochem. 108: 1012–1021, 2009.


American Journal of Physiology-renal Physiology | 2014

Rho-kinase regulation of TNF-α-induced nuclear translocation of NF-κB RelA/p65 and M-CSF expression via p38 MAPK in mesangial cells

Keiichiro Matoba; Daiji Kawanami; Masami Tsukamoto; Jun Kinoshita; Tomoko Ito; Sho Ishizawa; Yasushi Kanazawa; Tamotsu Yokota; Noriyuki Murai; Senya Matsufuji; Kazunori Utsunomiya

The small GTPase Rho and its downstream effector, Rho-associated coiled-coil containing protein kinase (Rho-kinase), regulate a number of cellular processes, including organization of the actin cytoskeleton, cell adhesion, and migration. While pharmacological inhibitors of Rho-kinase signaling are known to block renal inflammation, the molecular basis for this effect is unclear. Here, we provide evidence that proinflammatory TNF-α promotes mesangial expression of macrophage colony-stimulating factor (M-CSF), a key regulator for the growth and differentiation of mononuclear phagocytes, in a Rho-kinase-dependent manner. Consistent with this observation, TNF-α-mediated renal expression of M-CSF in insulin-resistant db/db mice was downregulated by Rho-kinase inhibition. Small interfering RNA-facilitated knockdown of Rho-kinase isoforms ROCK1 and ROCK2 indicated that both isoforms make comparable contributions to regulation of M-CSF expression in mesangial cells. From a mechanistic standpoint, Western blotting and EMSA showed that Rho-kinase and its downstream target p38 MAPK regulate nuclear translocation of NF-κB RelA/p65 and subsequent DNA binding activity, with no significant effects on IκBα degradation and RelA/p65 phosphorylation. Moreover, we showed that Rho-kinase-mediated cytoskeletal organization is required for the nuclear uptake of RelA/p65. Collectively, these findings identify Rho-kinase as a critical regulator of chemokine expression and macrophage proliferation.


Journal of Biological Chemistry | 1997

ATP-, K+-dependent Heptamer Exchange Reaction Produces Hybrids between GroEL and Chaperonin from Thermus thermophilus

Hideki Taguchi; Kei Amada; Noriyuki Murai; Mitsuko Yamakoshi; Masasuke Yoshida

Chaperonin from Thermus thermophilus(Tcpn6014· Tcpn107) splits at the plane between two Tcpn607 rings into two parts in a solution containing ATP and K+ (Ishii, N., Taguchi, H., Sasabe, H., and Yoshida, M. (1995) FEBS Lett. 362, 121–125). WhenEscherichia coli GroEL14 was additionally included in the solution described above, hybrid chaperonins GroEL7·Tcpn607 and GroEL7· Tcpn607·Tcpn107 were formed rapidly (<20 s) at 37u2009°C. The hybrid was also formed from Tcpn6014 and GroEL14 but not from a mutant GroEL14 lacking ATPase activity. The hybrid formation was saturated at ∼300 μm ATP and ∼300 mmK+. These results imply that GroEL14 also splits and undergoes a heptamer exchange reaction withThermus chaperonin under nearly physiological conditions. Similar to parent chaperonins, the isolated hybrid chaperonins exhibited ATPase activity that was susceptible to inhibition by Tcpn107 or GroES7 and mediated folding of other proteins. Once formed, the hybrid chaperonins were stable, and the parent chaperonins were not regenerated from the isolated hybrids under the same conditions in which the hybrids had been formed. Only under conditions in which GroEL in the hybrids was selectively destroyed, such as incubation at 70u2009°C, Thermus chaperonin, but not GroEL14, was regenerated from the hybrid. Therefore, the split reaction may not be an obligatory event repeated in each turnover of the chaperonin functional cycles but an event that occurs only when chaperonin is first exposed to ATP/K+.


BMC Cancer | 2014

MicroRNA-21 is a candidate driver gene for 17q23-25 amplification in ovarian clear cell carcinoma

Yukihiro Hirata; Noriyuki Murai; Nozomu Yanaihara; Misato Saito; Motoaki Saito; Mitsuyoshi Urashima; Yasuko Murakami; Senya Matsufuji; Aikou Okamoto

BackgroundEpithelial ovarian cancer (EOC) is the most common cause of gynecological malignancy-related mortality. Ovarian clear cell carcinoma (CCC) has unique clinical characteristics and behaviors that differ from other histological types of EOC, including a frequent association with endometriosis and a highly chemoresistant nature, resulting in poor prognosis. However, factors underlying its malignant behavior are still poorly understood. Aberrant expression of microRNAs has been shown to be involved in oncogenesis, and microRNA-21 (miR-21) is frequently overexpressed in many types of cancers. The aim of this study was to investigate the role of miR-21 in 17q23-25 amplification associated with CCC oncogenesis.MethodsWe identified 17q23-25 copy number aberrations among 28 primary CCC tumors by using a comparative genomic hybridization method. Next, we measured expression levels of the candidate target genes, miR-21 and PPM1D, for 17q23-25 amplification by real-time RT-PCR analysis and compared those data with copy number status and clinicopathological features. In addition, immunohistochemical analysis of PTEN (a potential target of miR-21) was performed using the same primary CCC cases. We investigated the biological significance of miR-21 overexpression in CCC using a loss-of-function antisense approach.Results17q23-25 amplification with both miR-21 overexpression and PTEN protein loss was detected in 4/28 CCC cases (14.2%). The patients with 17q23-25 amplification had significantly shorter progression-free and overall survival than those without 17q23-25 amplification (log-rank test: p = 0.0496; p = 0.0469, respectively). A significant correlation was observed between miR-21 overexpression and endometriosis. Both PTEN mRNA and PTEN protein expression were increased by miR-21 knockdown in CCC cells. We also confirmed that miR-21 directly bound to the 3′-untranslated region of PTEN mRNA using a dual-luciferase reporter assay.ConclusionsMiR-21 is a possible driver gene other than PPM1D for 17q23-25 amplification in CCC. Aberrant expression of miR-21 by chromosomal amplification might play an important role in CCC carcinogenesis through the regulation of the PTEN tumor suppressor gene.

Collaboration


Dive into the Noriyuki Murai's collaboration.

Top Co-Authors

Avatar

Senya Matsufuji

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Yasuko Murakami

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Masasuke Yoshida

Tokyo Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Hideki Taguchi

Tokyo Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Tomasz Hascilowicz

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Ayasa Tajima

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Jun Kinoshita

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Kazunori Utsunomiya

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Keiichiro Matoba

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Masami Tsukamoto

Jikei University School of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge