Network


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

Hotspot


Dive into the research topics where Jun-ichi Nishida is active.

Publication


Featured researches published by Jun-ichi Nishida.


Journal of Biological Chemistry | 1995

Mouse MTH1 Protein with 8-Oxo-7,8-dihydro-2′-deoxyguanosine 5′-Triphosphatase Activity That Prevents Transversion Mutation cDNA CLONING AND TISSUE DISTRIBUTION

Tetsuya Kakuma; Jun-ichi Nishida; Teruhisa Tsuzuki; Mutsuo Sekiguchi

8-Oxo-7,8-dihydro-2′-deoxyguanosine 5′-triphosphate (8-oxo-dGTP) is formed in the nucleotide pool of a cell during normal cellular metabolism, and when it is incorporated into DNA causes mutation. Organisms possess 8-oxo-dGTPase, an enzyme that specifically degrades 8-oxo-dGTP to 8-oxo-dGMP. We isolated cDNA for mouse 8-oxo-dGTPase, using as a probe human MTH1 (Escherichia coli mutT homolog) cDNA. The nucleotide sequence of the cDNA revealed that the mouse MTH1 protein (molecular weight of 17,896) comprises 156 amino acid residues. When the cDNA for mouse 8-oxo-dGTPase was expressed in E. coli mutT mutant cells devoid of their own 8-oxo-dGTPase activity, an 18-kDa protein, which is cross-reactive with an anti-human MTH1 antibody, was formed. In such cells, the level of spontaneous mutation frequency that was elevated reverted to normal. High levels of 8-oxo-dGTPase activity were found in liver, thymus, and large intestine, whereas all other organs examined contained smaller amounts of the enzyme. In embryonic stem cells, an exceedingly high level of the enzyme was present.


International Journal of Cancer | 2001

Sodium butyrate induces growth arrest and senescence‐like phenotypes in gynecologic cancer cells

Yasuhisa Terao; Jun-ichi Nishida; Shinji Horiuchi; Fengnian Rong; Yousuke Ueoka; Takao Matsuda; Hidenori Kato; Yoshiaki Furugen; Koyo Yoshida; Kiyoko Kato; Norio Wake

We demonstrated here the growth‐suppressing effects of sodium butyrate (NaB) on human endometrial and ovarian cancer cells. The arrest of cells at the G1 checkpoint accounted for this effect. NaB‐mediated p21 might arrest endometrial and ovarian cancer cells at the G0/G1 phase by eliciting pRb unphosphorylation. To demonstrate the role of pRb regulation by p21, we measured the sensitivity to NaB of cervical cancer cells in which pRb had been inactivated by HPV E7. The cervical cancer cells displayed a sensitivity in NaB‐mediated G2/M arrest in addition to their sensitivity in G0/G1 arrest. Arrest at G0/G1 and G2/M accompanied induction of senescence‐like phenotypes (SLPs). Most importantly, the effect of NaB on senescence induction was not coupled with the predominance of hypophosphorylated pRb forms in the cervical cancer cells. This suggested that NaB had the potential to elicit SLPs through p21‐mediated withdrawal from cell cycle progression. The consequences of p21 induction were manifold. The effects of NaB on gynecologic cancer cell growth indicated its potential use in cancer treatment. NaB was effective even in the cancer cells with mutant p53 and/or Rb genes by eliciting cell senescence.


British Journal of Cancer | 2000

Hepatocyte growth factor modulates motility and invasiveness of ovarian carcinomas via Ras-mediated pathway

Yousuke Ueoka; Kiyoko Kato; Yumiko Kuriaki; Shinji Horiuchi; Yasuhisa Terao; Jun-ichi Nishida; Hikaru Ueno; Norio Wake

Hepatocyte growth factor (HGF) is a multifunctional growth factor which has pleiotrophic biological effects on epithelial cells such as proliferation, motogenesis, invasiveness and morphogenesis. Peritoneal dissemination is critical for the progression of ovarian cancer, and our study revealed that HGF induces migration and invasion of ovarian cancer cells. We also demonstrated that HGF stimulates autophosphorylation of its receptor, followed by activation of the Ras-MAP (mitogen-activated peptide) kinase cascade. Moreover, infection of ovarian cancer cells with Ras dominant-negative adenovirus reduced the HGF-induced motogenic and invasive activities. Additionally, both MEK and PI3-kinase pathways downstream of Ras were involved in HGF-stimulated ovarian cancer cell invasiveness.


European Journal of Pharmaceutical Sciences | 2001

Photodynamic antisense regulation of human cervical carcinoma cell growth using psoralen-conjugated oligo(nucleoside phosphorothioate)

Akira Murakami; Asako Yamayoshi; Reiko Iwase; Jun-ichi Nishida; Tetsuji Yamaoka; Norio Wake

Abstract The antisense strategy has been applied to regulate gene expression in a sequence specific manner, which enables suppression of the proliferation of cancer cells and exploration of the functions of unknown genes. In order to generalize and to enhance the ability of the strategy, functionalization of antisense DNAs was done using a photo-crosslinking reagent, 4,5′,8-trimethylpsoralen, and the possibility of photodynamic antisense regulation of gene expression was examined. Psoralen-conjugated oligo(nucleoside phosphorothioate)s (Ps–S-oligo) were prepared and used to inhibit the proliferation of human cervical carcinoma cells. Upon UVA irradiation of Ps–S-oligo treated cells, Ps–S-oligo complementary to the initiation codon region (Ps–P-As) of HPV18-E6*-mRNA of human cervical carcinoma cells inhibited drastically the cell growth (IC 50 =16 nM). In contrast, Ps–S-oligo with mismatched sequences and scrambled one showed lesser inhibitory effects than Ps–P-As. These results showed that the inhibition by Ps–S-oligo was dependent on (a) sequence, (b) UVA irradiation, (c) concentration and (d) cell line. The amount of intact HPV18-E6*-mRNA was decreased in a sequence dependent manner, indicating that the antiproliferative effect of Ps–P-As was an antisense manner. The psoralen-conjugated antisense DNA has significant potential to regulate gene expression, which may provide useful information to explore the novel gene regulating reagents.


Cancer Research | 2004

K-Ras and H-Ras activation promote distinct consequences on endometrial cell survival.

Yumiko Ninomiya; Kiyoko Kato; Akira Takahashi; Yousuke Ueoka; Tetsuya Kamikihara; Takahiro Arima; Takao Matsuda; Hidenori Kato; Jun-ichi Nishida; Norio Wake

A considerable amount of evidence indicates that Ras signaling contributes to the development of endometrial cancer. We previously demonstrated that endometrial cancer cells carrying oncogenic [(12)Val]K-ras were susceptible to apoptosis. The present study examined the role of K-and H-Ras in the induction of apoptosis using rat endometrial cells (RENT4 cells). We found that constitutively activated K-Ras promoted apoptotic cell death, whereas the H-Ras mutant rescued rat endometrial cells from apoptosis. Expression of a constitutively active form of Raf-1 (Raf-CAAX) promoted apoptosis, whereas expression of a constitutively active catalytic subunit of phosphoinositide 3-kinase, p110K227E, allowed cells to escape from apoptosis. Moreover, inhibition of the MEK-MAPK pathway by the specific inhibitor, UO126, rescued the cells from apoptosis, whereas the inhibition of phosphoinositide 3-kinase by its specific inhibitor, LY294002, promoted apoptosis in RENT4 cells expressing activated K-Ras. However, both inhibitors promoted apoptosis in RENT4 cells expressing activated H-Ras. This difference in the regulation of apoptosis by the MEK inhibitor between K-Ras- and H-Ras-expressing cells depended on the interaction of effector proteins downstream of each Ras isoform. Finally, to elucidate the role of downstream K-Ras signal pathways, we generated K-Ras effector domain mutants (K12V35S, K12V40C). We examined the incidence of apoptotic cell death induced by the K-Ras effector domain mutants (K12V35S, K12V40C). The relative ratio of phospho-MAPK to phospho-Akt compared with that of mock cells was higher in K12V35S cells than in K12V40C cells. Ectopic expression of K12V35S protein increased the proportion of apoptotic cells, and in turn, the expression of K12V40C protein decreased compared with the expression of K12V protein without the effector domain mutant. These results demonstrate that K- and H-Ras-mediated signaling pathways exert distinct effects on apoptosis and that K-Ras downstream Raf/MEK/MAPK pathway is required for the induction of apoptosis in endometrial cells. Coordination of the two pathways contributes to endometrial cell survival.


Oncogene | 1997

Contribution of enhanced transcriptional activation by ER to [12Val] K-Ras mediated NIH3T3 cell transformation

Kiyoko Kato; Yousuke Ueoka; T. Hachiya; Jun-ichi Nishida; Norio Wake

We investigated the biological significance of estrogen receptors (ER) in NIH3T3 cell transformation by the [12Val] K-Ras mutant. This mutant enhanced the steady state level of ER. Cells expressing mutant K-Ras (K12V cell) were tumorigenic. To determine the role of ER accumulation in Ras-transformed cells, we developed cells (KwtER cells) that overexpressed both wild-type (wt) K-Ras and ER, and found these cells were also tumorigenic. E2 stimulated the transcriptional activity by ER dominantly in K12V cells. However, only partial activation of ER by E2 was seen in KwtER cells. In the presence of 10% serum in media, the activation of ER appeared only in transformed KtwER and K12V cells, suggesting that two independently transmitted signals, the E2-ER binding and the ER-AF1 activation, are necessary for ER activation and that the dominant activation of ER might be involved in Ras-mediated cell transformation. Co-expression of progesterone receptor (PR) with mutant K-Ras led to suppression of tumorigenicity and inhibition of the activation of ER. The antisense oligomers complementary to the ER suppressed proliferation and transformed phenotypes of K12V cells. These observations support the importance of ER in Ras-mediated cell transformation.


European Journal of Cancer | 1998

Oncogenic ras modulates epidermal growth factor responsiveness in endometrial carcinomas

Kiyoko Kato; Yousuke Ueoka; T Tamura; Jun-ichi Nishida; Norio Wake

Since the majority of endometrial carcinomas do not contain any detectable ras mutations, the precise contribution of aberrant Ras function, if any, to endometrial carcinoma development remains to be determined. Since there is considerable evidence that Ras transformation is associated with a decreased requirement for growth factors, we compared the growth response of endometrial carcinoma cells harbouring wild-type (Ishikawa cells) or mutated (HHUA cells) K-ras to epidermal growth factor (EGF). K-ras mutation did not significantly affect the level of the EGF receptor (EGFR) expressed in these carcinoma cells. EGF could stimulate the growth of Ishikawa, but not HHUA cells. Furthermore, EGF caused elevation of Ras-GTP levels in Ishikawa, but not HHUA cells. However, the introduction of mutated, but not normal, K-ras into Ishikawa cells rendered them non-responsive to EGF growth stimulation. Thus, the presence of mutated K-ras alone modulated the growth response of endometrial carcinoma cells to EGF. An inhibitor of the EGFR tyrosine kinase activity could prevent soft agar colony formation of Ishikawa cells, but not HHUA or mutant K-ras(12V)-transfected Ishikawa cells. Taken together, these results suggest that mutated K-ras causes a loss of responsiveness to EGF stimulation and that EGFR function is dispensable for the growth of mutant Ras-positive endometrial carcinoma cells.


Molecular Carcinogenesis | 1997

Alterations of the p16INK4A gene in human ovarian cancers

Tatsuya Kanuma; Jun-ichi Nishida; Tomonao Gima; J. Carl Barrett; Norio Wake

The p16INK4A gene, which encodes the cell‐cycle regulatory protein cyclin‐dependent kinase 4 inhibitor, is a putative tumor‐suppressor gene. We examined p16 gene alterations in 30 primary ovarian cancers and 11 ovarian cancer cell lines. Five of the primary cancers (16.7%) had lost both p16INK4A genes. In addition, four cancers (13.3%) contained five kinds of missense mutations and a one‐base deletion. Three cell lines had homozygous deletions of p16 genes, and one cell line had multiple intragenic mutations. There was also suppressed transcription of the p16 gene in one cell line. Some point mutations occurred in the conserved ankylin consensus region. These observations suggest that p16 is a functional target for ovarian carcinogenesis and that p16 alterations occurred in the primary cancers. Mol. Carcinog. 18:134–141, 1997.


Breast Cancer | 1999

Relevance of ER to the Development of Endometrial Hyperplasia and Adenocarcinoma.

Kiyoko Kato; Shinji Horiuchi; Yasuhisa Terao; Yousuke Ueoka; Jun-ichi Nishida; Daisuke Mori; Yasuji Yoshikawa; Norio Wake

Estrogen has an important role in both the etiology and treatment of hormone-dependent endometrial cancers, although the mechanism remains elusive. To define the role of estrogen-mediated signaling we investigated the biological significance of estrogen receptors (ER) in NIH3T3 cell transformation via the [12Val] K-Ras mutant. This mutant enhanced the steady state level and transcriptional activity of ER. In addition, overexpression of both wild type K-Ras and ER transformed NIH3T3 cells. Co-expression of the progesterone receptor (PR) with mutant K-Ras led to suppression of tumorigenicity and inhibition of ER activation. The antisense oligomers complementary to ER suppressed proliferation and transformed phenotypes of K12V cells. These observations support the importance of ER in Ras-mediated cell transformation.To address whether ER activation is also important in the development of human endometrial cancers, we investigated ER and PR expression levels in premalignant and malignant endometrial lesions. The results suggested the implication of ER abundance in endometrial hyperplasias, though modulation of PR expression by ER was retained. Gl adenocarcinoma also expressed higher levels of ER while PR modulation by ER was abrogated. These data implied the importance of ER activities in endometrial hyperplasia and Gl adenocarcinoma development.


Cancer Genetics and Cytogenetics | 1992

Chromosome alterations contribute to neoplastic progression of transformed rat embryonal fibroblasts

Hidenori Kato; Jun-ichi Nishida; Tsuyoshi Honda; Shingo Miyamoto; Kei Fujinaga; Norio Wake

Three types of transformants derived from rat embryonal fibroblasts (REFs) corresponding to the different progressional stages were obtained: TF1 (human papillomavirus type 16 E7 (HPV16 E7) transfection alone) and TF2 (E7 plus adenovirus type 12(Ad12) E1b were immortalized, TF3 (E7 plus adenovirus type 5 (Ad5) E1B) was anchorage-independent but not tumorigenic, and TF4 (E7 plus EJ-ras) was tumorigenic. Cytogenetic investigations revealed that the cells carrying specific chromosomal abnormalities expanded clonally in three of the five TF4 tumorigenic clones, in contrast to the TF1-TF3 non-tumorigenic clones, which showed a normal karyotype. By the inoculation of TF4 into syngeneic rats, 8 tumor-derived clones were obtained. Clonal expansion of cells carrying specific chromosome changes was also remarkable in these tumor-derived clones. However, the type of rearrangements and the chromosomes involved in the abnormalities were not identical. In addition, it was shown that chromosome constitutions of the parental TF4 transformants were apparently inconsistent with those of their tumor-derived clones. However, the clonal nature of abnormalities observed in the parental and the tumor-derived clones suggested that these genetic events of cellular genomes corresponded with and possibly contributed to the progression of malignant phenotypes of cells.

Collaboration


Dive into the Jun-ichi Nishida'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

Akira Murakami

Kyoto Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Asako Yamayoshi

Kyoto Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge