Network


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

Hotspot


Dive into the research topics where Taro Kanaya is active.

Publication


Featured researches published by Taro Kanaya.


Cancer Research | 2004

Human CYP1B1 Is Regulated by Estradiol via Estrogen Receptor

Yuki Tsuchiya; Miki Nakajima; Satoru Kyo; Taro Kanaya; Masaki Inoue; Tsuyoshi Yokoi

Human cytochrome P450 (CYP) 1B1 is a key enzyme in the metabolism of 17beta-estradiol (E2). CYP1B1 is mainly expressed in endocrine-regulated tissues, such as mammary, uterus, and ovary. Because many CYP enzymes are likely to be induced by the substrates themselves, we examined whether the human CYP1B1 expression is regulated by E2 in the present study. Real-time reverse transcription-PCR analysis revealed that treatment with 10 nM E2 for 12 h induced CYP1B1 mRNA expression in estrogen receptor (ER)-positive MCF-7 cells. Luciferase reporter assays using MCF-7 cells showed a significant transactivation up to 7-fold by E2 with a reporter plasmid containing a region from -152 to +25 of the human CYP1B1 gene. A computer-assisted homology search indicated a putative estrogen response element (ERE) between -63 and -49 in the CYP1B1 promoter region. Specific binding of ERalpha to the putative ERE was demonstrated by chromatin immunoprecipitation assays and gel shift analyses. With reporter plasmids containing the wild or mutated putative ERE on the CYP1B1 gene and the wild or mutated ERalpha expression vectors, luciferase assays using Ishikawa cells demonstrated that the putative ERE and ERalpha are essential for the transactivation by E2. Because endometrial tissue is highly regulated by estrogens, the expression pattern of CYP1B1 protein in human endometrial specimens was examined by immunohistochemistry. The staining of CYP1B1 was stronger in glandular epithelial cells during a proliferative phase than those during a secretory phase, consistent with the pattern of estrogen secretion. These findings clearly indicated that the human CYP1B1 is regulated by estrogen via ERalpha. Because 4-hydroxylation of estrogen by CYP1B1 leads to decrease of the estrogenic activity but the produced metabolite is toxicologically active, our findings suggest a clinical significance in the estrogen-regulated CYP1B1 expression for the homeostasis of estrogens as well as estrogen-dependent carcinogenesis.


American Journal of Pathology | 2003

Successful Immortalization of Endometrial Glandular Cells with Normal Structural and Functional Characteristics

Satoru Kyo; Mitsuhiro Nakamura; Tohru Kiyono; Yoshiko Maida; Taro Kanaya; Masaaki Tanaka; Noriyuki Yatabe; Masaki Inoue

The human endometrium is a dynamic tissue, the proliferative activity of which dramatically changes throughout the menstrual cycle, with exquisite regulation by sex-steroid hormones. Primary endometrial epithelial cells fall into senescence within 2 weeks when cultured on plastic dishes, and more complete understanding of endometrial biology has been delayed because of, in part, a lack of an in vitro culture model for endometrial epithelial cells. Our goal was to establish immortalized human endometrial glandular cells that retain the normal functions and characteristics of the primary cells. Because the Rb/p16 and p53 pathways are known to be critical elements of epithelial senescence in early passages, we used human papillomavirus E6/E7 to target these pathways. The combination of human papillomavirus-16 E6/E7 expression and telomerase activation by the introduction of human telomerase reverse transcriptase (hTERT) led to successful immortalization of the endometrial glandular cells. E6/E7 expression alone was sufficient to extend their life span more than 20 population doublings, but the telomerase activation was further required to enable the cells to pass through the subsequent replicative senescence at 40 population doublings. Isolated immortalized cells contained no chromosomal abnormalities or only nonclonal aberrations, retained responsiveness to sex-steroid hormones, exhibited glandular structure on three-dimensional culture, and lacked transformed phenotypes on soft agar or in nude mice. These findings support the notion that both Rb inactivation/p53 inactivation and telomerase activation are necessary to immortalize endometrial epithelial cells, but additional factors are required for endometrial carcinogenesis. Our established cell lines show great promise for investigation of hormone functions, endometrial biology, and endometrial carcinogenesis.


International Journal of Cancer | 1999

Human telomerase reverse transcriptase as a critical determinant of telomerase activity in normal and malignant endometrial tissues

Satoru Kyo; Taro Kanaya; Masahiro Takakura; Masaaki Tanaka; Masaki Inoue

Telomerase activation is thought to be essential for cellular immortality and oncogenesis. It is observed in most malignant tumors but not in most normal somatic tissues. Normal human endometrium is, however, known to express significant telomerase activity in a menstrual phase-dependent manner. The 3 major subunits composing telomerase have been identified. Using normal and malignant endometrial tissues, we studied how these components are involved in telomerase activation. A total of 23 endometrial cancers and 32 normal human endometria in various menstrual phases as well as cell lines derived from endometrial cancer were examined for the expression of each telomerase subunit using RT-PCR analysis. Telomerase activity in each sample was determined by the TRAP assay, and the correlation between subunit expression and telomerase activity was examined. RT-PCR analysis revealed that telomerase RNA (hTR) and telomerase-associated protein (TP1) mRNA were constitutively expressed in both normal and malignant endometrial tissues. Expression of human telomerase reverse transcriptase (hTERT) mRNA was observed in most endometrial cancers, while that in normal endometrium depended on the phases of menstrual cycles. Proliferative phase normal endometria expressed hTERT mRNA, while secretory phase endometria did not. There was a strong association between telomerase activity and hTERT expression but not TP1 or hTR expression in both normal and tumor tissues. Five telomerase-positive endometrial cancer cell lines expressed each of the telomerase subunits including hTERT, while 2 telomerase-negative normal primary fibroblast cells expressed TP1 mRNA and hTR, but not hTERT mRNA. Our findings suggest that hTERT is a rate-limiting determinant of enzymatic activity of human telomerase. Since some normal tissues with high regenerative potential can express hTERT, special attention should be paid to the clinical use of hTERT inhibitors as anti-cancer drugs.


Oncogene | 2002

Direct activation of telomerase by EGF through Ets-mediated transactivation of TERT via MAP kinase signaling pathway

Yoshiko Maida; Satoru Kyo; Taro Kanaya; Zhuo Wang; Noriyuki Yatabe; Masaaki Tanaka; Mitsuhiro Nakamura; Masahide Ohmichi; Noriko Gotoh; Seishi Murakami; Masaki Inoue

Telomerase is a regulated enzyme and its activity is tightly associated with cell proliferation. The mechanisms of this association are unclear, but specific growth factors may regulate telomerase activity. The present study examines the effect of epidermal growth factor (EGF) on telomerase activity and identifies the signal transduction pathway involved in this process. EGF up-regulated telomerase activity in EGF receptor-positive cells after the activation of telomerase reverse transcriptase (TERT) mRNA expression. This activation was rapid, peaked after 6 or 12 h and was not blocked by the concurrent exposure to cycloheximide, suggesting a direct effect of EGF on TERT transcription. Transient expression assays revealed that EGF activates the hTERT promoter and that the proximal core promoter is responsible for this regulation. The activation of hTERT mRNA expression by EGF was specifically blocked by MEK inhibitor, and in vitro kinase assays demonstrated that ERK is activated in response to EGF. Transient expression assays using mutant reporter plasmids revealed that an ETS motif located in the core promoter of hTERT is required for the EGF-induced transactivation of hTERT. Overexpression of wild type Ets in cells enhanced the EGF effect on hTERT transcription, while that of dominant negative Ets significantly repressed EGF action. These findings suggest that EGF activates telomerase through the direct activation of TERT transcription, in which the Ras/MEK/ERK pathway and Ets factor play major roles. Our data support the notion that growth factors directly regulate telomerase via specific signal transduction pathways.


Nucleic Acids Research | 2000

Identification and characterization of negative regulatory elements of the human telomerase catalytic subunit (hTERT) gene promoter: possible role of MZF-2 in transcriptional repression of hTERT

Kohtaro Fujimoto; Satoru Kyo; Masahiro Takakura; Taro Kanaya; Yasuhide Kitagawa; Hideaki Itoh; Morinobu Takahashi; Masaki Inoue

Human telomerase reverse transcriptase (hTERT) is a catalytic subunit of human telomerase and is a critical determinant of the enzymatic activity of telomerase. Expression of hTERT is known to be regulated mainly at the transcriptional level. In the present study, using transient expression assays, we identified a 400 bp silencer of the hTERT promoter between -776 and -378 upstream of the proximal core promoter. The inhibitory effects of this silencer were enhanced with cellular differentiation. A computer-assisted homology search identified multiple binding motifs for myeloid-specific zinc finger protein 2 (MZF-2) within this region. Mutation introduced in these sites resulted in significant activation of hTERT transcription. Gel shift assays demonstrated that MZF-2 proteins specifically bound to these sites. Overexpression of MZF-2 in cells led to down-regulation of hTERT transcription as well as telomerase activity. These findings suggest that the 400 bp region upstream of the hTERT core promoter that we identified functions as a negative regulatory region and that MZF-2 may be an effector of negative regulation of hTERT.


International Journal of Cancer | 1998

hTERT is a critical determinant of telomerase activity in renal-cell carcinoma

Taro Kanaya; Satoru Kyo; Masahiro Takakura; Hideaki Ito; Mikio Namiki; Masaki Inoue

Telomerase is a ribonucleoprotein enzyme which stabilizes chromosomal structure, thereby inducing cellular immortality. Three major subunits composing telomerase complex have been cloned, designated hTR (human telomerase RNA), TP1 (telomerase‐associated protein 1), and hTERT (human telomerase reverse transcriptase). In the present study, a total of 36 renal‐cell carcinomas(RCC) and adjacent normal tissues, as well as cell lines derived from RCC or normal kidney, were examined for the expression of each telomerase subunit and telomerase activity. RT‐PCR analyses revealed that hTR and TP1 mRNA were constitutively expressed both in tumor and in normal tissues. In contrast, hTERT mRNA was expressed in most tumors, but not in normal tissues. Telomeric‐repeat‐amplification‐protocol (TRAP) assay revealed that more than 80% of RCC tumor tissue exhibited telomerase activity, while none of the adjacent normal tissue did. There was a significant association of telomerase activity with expression of hTERT mRNA, but not with TP1 mRNA or hTR expression. Two cell lines, derived from RCC and cervical cancer, expressed telomerase activity and hTERT mRNA, while normal renal cortical epithelial cells expressed neither of them. These findings suggest that hTERT plays a critical role in determining the enzymatic activity of human telomerase, and that up‐regulation of hTERT probably plays a role in the progression of human cancers. Int. J. Cancer 78:539–543, 1998.


Oncogene | 2004

HIF-1-mediated activation of telomerase in cervical cancer cells

Noriyuki Yatabe; Satoru Kyo; Yoshiko Maida; Hirotaka Nishi; Mitsuhiro Nakamura; Taro Kanaya; Masaaki Tanaka; Keiichi Isaka; Satoshi Ogawa; Masaki Inoue

Hypoxia-inducible factor 1 (HIF-1) is a key regulator of O2 homeostasis, which regulates the expression of several genes linked to angiogenesis and energy metabolism. Tumor hypoxia has been shown to be associated with poor prognosis in a variety of tumors, and HIF-1 induced by hypoxia plays pivotal roles in tumor progression. The presence of putative HIF-1-binding sites on the promoter of human telomerase reverse transcriptase gene (hTERT) prompted us to examine the involvement of HIF-1 in the regulation of hTERT and telomerase in tumor hypoxia. The telomeric repeat amplification protocol (TRAP) assay revealed that hypoxia activated telomerase in cervical cancer ME180 cells, with peak induction at 24–48 h of hypoxia. Notably, hTERT mRNA expression was upregulated at 6–12 h of hypoxia, concordant with the elevation of HIF-1 protein levels at 6 h. hTERT protein levels were subsequently upregulated at 24 h and later. Luciferase assays using reporter plasmids containing hTERT core promoter revealed that hTERT transcription was significantly activated in hypoxia and by HIF-1 overexpression, and that the two putative binding sites within the core promoter are responsible for this activation. Chromatin immunoprecipitation assay identified the specific binding of HIF-1 to these sites (competing with c-Myc), which was enhanced in hypoxia. The present findings suggest that hypoxia activates telomerase via transcriptional activation of hTERT, and that HIF-1 plays a critical role as a transcription factor. They also suggest the existence of novel mechanisms of telomerase activation in cancers, and have implications for the molecular basis of hypoxia-induced tumor progression and HIF-1-based cancer gene therapy.


International Journal of Cancer | 1999

Expression of human telomerase subunits in ovarian malignant, borderline and benign tumors.

Satoru Kyo; Taro Kanaya; Masahiro Takakura; Masaaki Tanaka; Atsuko Yamashita; Hirokazu Inoue; Masaki Inoue

Telomerase activity is involved in the maintenance of telomere length and is thought to be required for cellular immortality and oncogenesis. Three major subunits composing telomerase, human telomerase RNA (hTR), telomerase‐associated protein (TP1) and human telomerase catalytic subunit (hTERT), have been identified. However, their functions and the regulatory mechanisms by which telomerase is activated have not been fully determined. In the present study, a total of 35 epithelial ovarian cancers, 5 ovarian low potential malignancies (LPM), 11 ovarian benign cysts and 12 normal ovaries, as well as various cell lines derived from ovarian cancers, were examined for the expression of hTR, TP1 mRNA and hTERT mRNA. Correlations of expression with telomerase activity were evaluated. Reverse transcription‐polymerase chain reaction (RT‐PCR) analysis revealed that hTR and TP1 mRNA were expressed in more than 80% of ovarian cancers, LPM, ovarian cysts and even in normal ovaries. However, hTERT mRNA was observed only in ovarian cancers, most of which exhibited telomerase activity. Normal ovarian tissues, ovarian cysts and LPM, most of which had no telomerase activity, did not express hTERT. Five telomerase‐positive ovarian cancer cell lines expressed each of the telomerase subunits, whereas 2 telomerase‐negative normal primary fibroblast cell lines expressed TP1 mRNA and hTR, but not hTERT mRNA. There was a significant correlation of telomerase activity with hTERT mRNA expression but not with TP1 or hTR expression. Expression of hTERT is thus specific to cancer lesions and appears to be a rate‐limiting determinant of the enzymatic activity of human telomerase. Up‐regulation of hTERT may play a critically important role in the development of ovarian cancers. Int. J. Cancer 80:804–809, 1999.


Oncogene | 2003

Frequent hypermethylation of MLH1 promoter in normal endometrium of patients with endometrial cancers

Taro Kanaya; Satoru Kyo; Yoshiko Maida; Noriyuki Yatabe; Masaaki Tanaka; Mitsuhiro Nakamura; Masaki Inoue

Silencing of the MLH1 gene by promoter hypermethylation is the mechanism underlying the microsatellite instability (MSI) phenotype in endometrial cancers. However, the profile of CpG methylation in a wide range of MLH1 promoters in endometrial cancers and in the normal endometrium is largely unknown. The present study investigates the region 700 bp upstream of MLH1 covering 48 CpG sites using bisulfite sequencing. Methylation status was classified as full (over 80% of CpGs are methylated), partial (10–80%) or nonmethylation (less than 10%). Of 56 endometrioid endometrial cancers, 16 (29%) were fully methylated, 14 (25%) were partially methylated and 26 (46%) were Q1not methylated. Analyses of MLH1 by immunohistochemical means and of MSI revealed that the Q2degree, rather than region-specific methylation of CpG islands is critical for decreased MLH1 expression and the MSI phenotype. Among 12 patients with methylated cancers, five (42%) patients contained methylated promoters in their normal endometria with profiles similar to those of cancer lesions, and these were associated with the MSI phenotype. In contrast, only one of 31 (3%) normal endometria from patients without endometrial malignancies harbored methylated promoters. These findings suggest that hypermethylation of the MLH1 promoter is frequent in the histologically normal endometrium adjacent to cancers, supporting the notion that hypermethylation of mismatch repair genes is the initial step that triggers various genetic events in endometrial carcinogenesis.


American Journal of Pathology | 1998

Expression of Telomerase Activity in Human Endometrium Is Localized to Epithelial Glandular Cells and Regulated in a Menstrual Phase- Dependent Manner Correlated with Cell Proliferation

Masaaki Tanaka; Satoru Kyo; Masahiro Takakura; Taro Kanaya; Tetsuya Sagawa; Kaname Yamashita; Yasunori Okada; Eiso Hiyama; Masaki Inoue

Telomerase activity is observed in most malignant tumors and germ cells, whereas normal somatic cells usually do not express it. Human endometrium is composed of glandular and stromal components and exhibits dramatic changes in proliferative activity during the menstrual cycle, which is exquisitely regulated by estrogen function. We previously reported that normal human endometrium expresses telomerase activity. However, it remains unclear which of the above components are the major sources of telomerase activity and how levels of telomerase activity are regulated over the menstrual cycle. Quantitative analysis of telomerase activity revealed that it changes dramatically over the course of the menstrual cycle and is strictly regulated in a menstrual-phase-dependent manner. Maximal activity equivalent to that in endometrial cancer was present in late proliferative phase, and minimal activity in late secretory phase. Postmenopausal endometrium and endometrium treated with anti-estrogen drugs exhibited decreased telomerase activity. Testing isolated epithelial glandular cells and stromal cells, we found that telomerase activity was localized to epithelial glandular cells. In situ RNA hybridization analysis also revealed epithelial-specific expression of human telomerase RNA. In vitro analysis of cultured epithelial cells demonstrated that telomerase activity is correlated with epithelial proliferation but not affected by estrogen treatment. These findings suggest that expression of telomerase activity is specific to epithelial cells and linked to cell proliferative status. The involvement of estrogen in telomerase regulation remains to be elucidated.

Collaboration


Dive into the Taro Kanaya'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