Network


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

Hotspot


Dive into the research topics where Norihisa Uehara is active.

Publication


Featured researches published by Norihisa Uehara.


International Journal of Oncology | 2012

Resveratrol induces apoptosis via ROS-triggered autophagy in human colon cancer cells

Hisanori Miki; Norihisa Uehara; Ayako Kimura; Tomo Sasaki; Takashi Yuri; Katsuhiko Yoshizawa; Airo Tsubura

Resveratrol (Res; 3,4′,5-trihydroxy-trans-stilbene), which is a polyphenol found in grapes, can block cell proliferation and induce growth arrest and/or cell death in several types of cancer cells. However, the precise mechanisms by which Res exerts anticancer effects remain poorly understood. Res blocked both anchorage-dependent and -independent growth of HT-29 and COLO 201 human colon cancer cells in a dose- and time-dependent manner. Annexin V staining and Western blot analysis revealed that Res induced apoptosis accompanied by an increase in Caspase-8 and Caspase-3 cleavage. In HT-29 cells, Res caused autophagy as characterized by the appearance of autophagic vacuoles by electron microscopy and elevation of microtubule-associated protein 1 light chain 3 (LC3)-II by immunoblotting, which was associated with the punctuate pattern of LC3 detected by fluorescein microscopy. Inhibition of Res-induced autophagy by the autophagy inhibitor 3-methyladenine caused a significant decrease in apoptosis accompanied by decreased cleavage of Casapse-8 and Caspase-3, indicating that Res-induced autophagy was cytotoxic. However, inhibition of Res-induced apoptosis by the pan-caspase inhibitor Z-VAD(OMe)-FMK did not decrease autophagy but elevated LC3-II levels. Interestingly, Res increased the intracellular reactive oxygen species (ROS) level, which correlated to the induction of Casapse-8 and Caspase-3 cleavage and the elevation of LC3-II; treatment with ROS scavenger N-acetyl cysteine diminished this effect. Therefore, the effect of Res on the induction of apoptosis via autophagy is mediated through ROS in human colon cancer cells.


Anti-cancer Agents in Medicinal Chemistry | 2011

Anticancer Effects of Garlic and Garlic-derived Compounds for Breast Cancer Control

Airo Tsubura; Yen Chang Lai; Maki Kuwata; Norihisa Uehara; Katsuhiko Yoshizawa

Garlic and garlic-derived compounds reduce the development of mammary cancer in animals and suppress the growth of human breast cancer cells in culture. Oil-soluble compounds derived from garlic, such as diallyl disulfide (DADS), are more effective than water-soluble compounds in suppressing breast cancer. Mechanisms of action include the activation of metabolizing enzymes that detoxify carcinogens, the suppression of DNA adduct formation, the inhibition of the production of reactive oxygen species, the regulation of cell-cycle arrest and the induction of apoptosis. Selenium-enriched garlic or organoselenium compounds provide more potent protection against mammary carcinogenesis in rats and greater inhibition of breast cancer cells in culture than natural garlic or the respective organosulfur analogues. DADS synergizes the effect of eicosapentaenoic acid, a breast cancer suppressor, and antagonizes the effect of linoleic acid, a breast cancer enhancer. Moreover, garlic extract reduces the side effects caused by anti-cancer agents. Thus, garlic and garlic-derived compounds are promising candidates for breast cancer control.


Molecular Cancer Research | 2008

Mesothelin Promotes Anchorage-Independent Growth and Prevents Anoikis via Extracellular Signal-Regulated Kinase Signaling Pathway in Human Breast Cancer Cells

Norihisa Uehara; Yoichiro Matsuoka; Airo Tsubura

Mesothelin (MSLN) is a glycoprotein that is overexpressed in various tumors. MSLN is present on the cell surface and is also released into body fluids or culture supernatants from MSLN-positive tumor cells. Despite intensive study of MSLN as a diagnostic marker or target for immunotherapy, its biological function is largely unknown. In the present study, we examined the effects of ectopic expression of MSLN in human breast cancer cell lines (MCF-7, T47D, and MDA-MB-231). We found that overexpression of MSLN promoted anchorage-independent growth in soft agar. In addition, MDA-MB-231 cells expressing high levels of MSLN exhibited resistance to anoikis (a type of apoptosis induced by detachment from substratum), as indicated by decreased DNA fragmentation and down-regulation of the proapoptotic protein Bim. Incubating MSLN-expressing MDA-MB-231 cells in the presence of U0126, an inhibitor of mitogen-activated protein/extracellular-signal-regulated kinase kinase, induced accumulation of Bim and restored susceptibility to anoikis. Western blot analysis also revealed that overexpression of MSLN resulted in sustained activation of extracellular signal-regulated kinase 1/2 and suppression of Bim. The present results constitute novel evidence that MSLN enables cells to survive under anchorage-independent conditions by suppressing Bim induction via the extracellular signal-regulated kinase signaling pathway. (Mol Cancer Res 2008;6(2):186–93)


Oncogene | 2007

TAK1 represses transcription of the human telomerase reverse transcriptase gene

Tsukasa Fujiki; Takumi Miura; M. Maura; H. Shiraishi; S Nishimura; Y Imada; Norihisa Uehara; Kosuke Tashiro; Sanetaka Shirahata; Yoshinori Katakura

In human cells, telomerase activity is tightly regulated by the expression of its catalytic subunit, namely, the human telomerase reverse transcriptase (hTERT). However, the molecular mechanisms involved in the regulation of hTERT expression have not been completely clarified. We have previously reported that transforming growth factor β (TGF-β) represses the expression of the hTERT gene. In the present study, we demonstrated that TGF-β-activated kinase 1 (TAK1), originally identified as a mitogen-activated kinase kinase kinase, represses the hTERT core promoter activity in an E-box-independent manner, and it also represses the transcription of the hTERT gene in human lung adenocarcinoma cell line, A549 cells. This TAK1-induced repression was found to be caused by the recruitment of histone deacetylase to Sp1 at the hTERT promoter and a consequent reduction in the amount of acetylated histone H4 at the hTERT promoter. Finally, we demonstrated that TAK1 induces cellular senescence programs in normal human diploid cells. Thus, we assume that TAK1 triggers the repression mechanisms of the hTERT gene as a result of evoking cellular senescence programs. Considered together, TAK1 is thought to play a causative role in the determination of a finite replicative lifespan of normal and cancer cells.


Journal of Mammary Gland Biology and Neoplasia | 2005

Dietary factors modifying breast cancer risk and relation to time of intake.

Airo Tsubura; Norihisa Uehara; Yasuhiko Kiyozuka; Nobuaki Shikata

Multiple factors contribute to the development of human breast cancer. However, environmental factors, especially dietary factors, appear to have the greatest effects. Evidence obtained in epidemiological studies has been corroborated by laboratory findings. Dietary components strongly associated with breast cancer include fat and phytochemicals. A diet high in n-3 polyunsaturated fatty acid (PUFA) or monounsaturated fatty acid (MUFA) and low in n-6 PUFA is protective against breast cancer. Some phytochemicals present in fruits and vegetables are also protective. Time of intake appears to be important: lifetime protection may be achieved if one is exposed to a dietary factor that lowers breast cancer risk early in life. Synergistic and antisynergistic interactions between dietary factors can modify breast cancer risk. The available evidence suggests that breast cancer risk can be reduced by early dietary intervention.


Breast Cancer Research | 2004

Conjugated docosahexaenoic acid suppresses KPL-1 human breast cancer cell growth in vitro and in vivo: potential mechanisms of action

Miki Tsujita-Kyutoku; Takashi Yuri; Naoyuki Danbara; Hideto Senzaki; Yasuhiko Kiyozuka; Norihisa Uehara; Hideho Takada; Takahiko Hada; Teruo Miyazawa; Yutaka Ogawa; Airo Tsubura

IntroductionThe present study was conducted to examine the effect of conjugated docosahexaenoic acid (CDHA) on cell growth, cell cycle progression, mode of cell death, and expression of cell cycle regulatory and/or apoptosis-related proteins in KPL-1 human breast cancer cell line. This effect of CDHA was compared with that of docosahexaenoic acid (DHA).MethodsKPL-1 cell growth was assessed by colorimetric 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay; cell cycle progression and mode of cell death were examined by flow cytometry; and levels of expression of p53, p21Cip1/Waf1, cyclin D1, Bax, and Bcl-2 proteins were examined by Western blotting analysis. In vivo tumor growth was examined by injecting KPL-1 cells subcutaneously into the area of the right thoracic mammary fat pad of female athymic mice fed a CDHA diet.ResultsCDHA inhibited KPL-1 cells more effectively than did DHA (50% inhibitory concentration for 72 hours: 97 μmol/l and 270 μmol/l, respectively). With both CDHA and DHA growth inhibition was due to apoptosis, as indicated by the appearance of a sub-G1 fraction. The apoptosis cascade involved downregulation of Bcl-2 protein; Bax expression was unchanged. Cell cycle progression was due to G0/G1 arrest, which involved increased expression of p53 and p21Cip1/Waf1, and decreased expression of cyclin D1. CDHA modulated cell cycle regulatory proteins and apoptosis-related proteins in a manner similar to that of parent DHA. In the athymic mouse system 1.0% dietary CDHA, but not 0.2%, significantly suppressed growth of KPL-1 tumor cells; CDHA tended to decrease regional lymph node metastasis in a dose dependent manner.ConclusionCDHA inhibited growth of KPL-1 human breast cancer cells in vitro more effectively than did DHA. The mechanisms of action involved modulation of apoptosis cascade and cell cycle progression. Dietary CDHA at 1.0% suppressed KPL-1 cell growth in the athymic mouse system.


Laboratory Investigation | 2014

Mesenchymal stem cells markedly suppress inflammatory bone destruction in rats with adjuvant-induced arthritis

Toshio Takano; Yin Ji Li; Akiko Kukita; Takayoshi Yamaza; Yasunori Ayukawa; Kanako Moriyama; Norihisa Uehara; Hisayuki Nomiyama; Kiyoshi Koyano; Toshio Kukita

Mesenchymal stem cells (MSCs) have potential to differentiate into multiple cell lineages. Recently, it was shown that MSCs also have anti-inflammatory and immunomodulatory functions. In this report, we investigated the regulatory function of MSCs in the development of inflammatory bone destruction in rats with adjuvant-induced arthritis (AA rats). MSCs were isolated from rat bone marrow tissues, expanded in the presence of basic FGF, and intraperitoneally injected into AA rats. MSC administration significantly suppressed inflammatory parameters: swelling score, swelling width, and thickness of hind paw. Radiographic evaluation indicated that MSC significantly suppressed bone destruction. Histological analysis showed that administration of MSCs markedly suppressed osteoclastogenesis in AA rats. To further delineate their effects on osteoclastogenesis, MSCs were added to in vitro bone marrow cultures undergoing osteoclastogenesis. MSCs significantly suppressed osteoclastogenesis in this system. Chemokine receptor expression in MSCs was assessed by RT-PCR, and a chemotactic assay was performed using a transwell culture system. MSCs showed significant chemotaxis to MIP-1α (CCL3) and SDF-1α (CXCL12), chemokines preferentially expressed in the area of inflammatory bone destruction. Furthermore, MSCs expressed IL-10 and osteoprotegerin, cytokines that suppress osteoclastogenesis. These data suggest that recruitment of MSC to the area of bone destruction in AA rats could suppress inflammatory bone destruction and raise the possibility that MSCs may have potential for the treatment of inflammatory bone destruction in arthritis.


Breast Cancer Research | 2003

Id-1 is not expressed in the luminal epithelial cells of mammary glands

Norihisa Uehara; Yu-Chien Chou; Jose J. Galvez; Paola de-Candia; Robert D. Cardiff; Robert Benezra; G. Shyamala

BackgroundThe family of inhibitor of differentiation/DNA binding (Id) proteins is known to regulate development in several tissues. One member of this gene family, Id-1, has been implicated in mammary development and carcinogenesis. Mammary glands contain various cell types, among which the luminal epithelial cells are primarily targeted for proliferation, differentiation and carcinogenesis. Therefore, to assess the precise significance of Id-1 in mammary biology and carcinogenesis, we examined its cellular localization in vivo using immunohistochemistry.MethodsExtracts of whole mammary glands from wild type and Id-1 null mutant mice, and tissue sections from paraffin-embedded mouse mammary glands from various developmental stages and normal human breast were subjected to immunoblot and immunohistochemical analyses, respectively. In both these procedures, an anti-Id-1 rabbit polyclonal antibody was used for detection of Id-1.ResultsIn immunoblot analyses, using whole mammary gland extracts, Id-1 was detected. In immunohistochemical analyses, however, Id-1 was not detected in the luminal epithelial cells of mammary glands during any stage of development, but it was detected in vascular endothelial cells.ConclusionId-1 is not expressed in the luminal epithelial cells of mammary glands.


Cancer Letters | 2012

Requirement of p38 MAPK for a cell-death pathway triggered by vorinostat in MDA-MB-231 human breast cancer cells

Norihisa Uehara; Sayaka Kanematsu; Hisanori Miki; Katsuhiko Yoshizawa; Airo Tsubura

Vorinostat is a histone deacetylase inhibitor that effectively suppresses cancer-cell proliferation by inducing cell-cycle arrest and/or apoptosis. We now show the involvement of p38 mitogen-activated protein kinase (MAPK) in the regulation of vorinostat-induced apoptosis in MDA-MB-231 human breast cancer cells. Vorinostat induced the hyperacetylation of histone H3, which correlated to apoptosis induction. Vorinostat-induced apoptosis occurred in parallel with the phosphorylation of p38 MAPK and the dephosphorylation of extracellular signal-regulated kinases 1 and 2 (ERK1/2). Knockdown of p38 MAPK prominently abrogated apoptosis induction and was accompanied by decreased caspase-3 cleavage. These findings support the notion that the activation of the p38 MAPK pathway followed by caspase-3 cleavage is responsible for vorinostat-induced apoptosis in MDA-MB-231 cells.


Medical Molecular Morphology | 2007

Multistep mouse mammary tumorigenesis through preneoplasia to neoplasia and acquisition of metastatic potential

Airo Tsubura; Katsuhiko Yoshizawa; Norihisa Uehara; Takashi Yuri; Yoichiro Matsuoka

Human breast tissue can give rise to hyperplasias, atypical hyperplasias, and in situ carcinomas originating in a terminal duct-lobular unit (TDLU). These entities are associated with increased risk of subsequent development of invasive carcinoma. Human breast carcinomas arise via intermediate steps known as precursor or premalignant lesions. However, it is difficult to perform stepwise observation of the progression of human breast cancer. Mouse mammary tissue can give rise to several characteristic types of premalignant hyperplasia and tumor, originating in a duct or acinus, that progress to carcinoma. Three specific types of mouse mammary lesion with premalignant potential have been identified: hyperplastic alveolar nodule (HAN), plaque (PLQ), and ductal hyperplasia (DH). These lesions partially resemble human precursor lesions, and clarification of similarities and differences between the human and mouse lesions could improve our understanding of human breast carcinogenesis. Some invasive breast carcinomas acquire metastatic potential and may cause the death of the patient. Because the most malignant aspect of neoplasia is metastasis, recognition of metastatic phenotypes is particularly important. Mouse mammary carcinomas rarely metastasize, but there exist mouse models of metastasis of mammary carcinoma. In the present review article, we describe morphological, biological, and molecular aspects of premalignant lesions of mice, and we discuss their metastatic phenotypes.

Collaboration


Dive into the Norihisa Uehara's collaboration.

Top Co-Authors

Avatar

Airo Tsubura

Kansai Medical University

View shared research outputs
Top Co-Authors

Avatar

Takashi Yuri

Kansai Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hisanori Miki

Kansai Medical University

View shared research outputs
Top Co-Authors

Avatar

Ayako Kimura

Kansai Medical University

View shared research outputs
Top Co-Authors

Avatar

Nobuaki Shikata

Kansai Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tomo Sasaki

Kansai Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge