Network


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

Hotspot


Dive into the research topics where Katsuhiro Uzawa is active.

Publication


Featured researches published by Katsuhiro Uzawa.


British Journal of Cancer | 2013

MicroRNA-125b regulates proliferation and radioresistance of oral squamous cell carcinoma.

Masashi Shiiba; Keiji Shinozuka; Kengo Saito; Kazuaki Fushimi; Atsushi Kasamatsu; Katsunori Ogawara; Katsuhiro Uzawa; Hiroshi Ito; Yuichi Takiguchi; Hideki Tanzawa

Background:MicroRNAs (miRNAs) are involved in essential biological activities, and have been reported to exhibit differential expression profiles in various cancers. Our previous study demonstrated that intercellular adhesion molecule-2 (ICAM2) inhibition induces radiosensitisation in oral squamous cell carcinoma (OSCC) cells. Thus, we hypothesised that certain miRNAs play crucial roles in radioresistance in OSCC by regulating ICAM2 expression.Methods:Because predicted target gene analyses revealed that microRNA-125b (miR-125b) potentially regulates ICAM2 mRNA expression, we examined the association between miR-125b and radioresistance. The expression of miR-125b was investigated by real-time quantitative reverse transcriptase–PCR. For a functional analysis, miR-125b was transfected to OSCC-derived cells.Results:A downregulated expression of miR-125b was found in OSCC-derived cell lines and OSCC samples. The miR-125b-transfected cells showed a decreased proliferation rate, enhanced radiosensitivity to X-ray irradiation and diminished ICAM2 mRNA expression. Moreover, miR-125b expression correlated with OSCC tumour staging and survival.Conclusion:These findings suggested that the downregulated miR-125b expression was associated with proliferation and radioresistance mechanisms, probably through ICAM2 signalling. Thus, controlling the expression or activity of miR-125b might contribute to suppressing proliferation and overcoming radioresistance in OSCC.


PLOS ONE | 2013

Overexpression of CDCA2 in Human Squamous Cell Carcinoma: Correlation with Prevention of G1 Phase Arrest and Apoptosis

Fumihiko Uchida; Katsuhiro Uzawa; Atsushi Kasamatsu; Hiroaki Takatori; Yosuke Sakamoto; Katsunori Ogawara; Masashi Shiiba; Hiroki Bukawa; Hideki Tanzawa

Cell division cycle associated 2 (CDCA2) recruits protein phosphatase 1 to chromatin to antagonize activation of ataxia telangiectasia mutated (ATM)-dependent signal transduction. ATM kinase plays a critical role in the DNA damage response and its phosphorylation cascade to inhibit the p53-MDM2 interaction, which releases p53 to induce p21 and G1 cell-cycle arrest. However, the relevance of CDCA2 to human malignancy including oral squamous cell carcinoma (OSCC) is unknown. In the current study, we found that CDCA2 expression was up-regulated in OSCC cell lines. Functional studies with shRNA system showed that knockdown of CDCA2 significantly (P<0.05) inhibited cellular proliferation compared with the control cells by arresting cell-cycle progression at the G1 phase and up-regulating the cyclin-dependent kinase inhibitors (p21Cip1, p27Kip1, p15INK4B, and p16INK4A). CDCA2 knockdown also promoted apoptosis after treatment with the DNA damage reagent, cisplatin. In clinical samples, the CDCA2 protein expression level in primary OSCCs was significantly (P<0.05) greater than in matched normal oral tissues (67/85, 79%). Furthermore, CDCA2-positive cases were correlated significantly (P<0.05) with high cancer progression. Our results showed for the first time that CDCA2 frequently is overexpressed in OSCCs and might be associated closely with OSCC progression by preventing cell-cycle arrest and apoptosis.


PLOS ONE | 2013

Kinesin Family member 4A: A Potential Predictor for Progression of Human Oral Cancer

Yasuyuki Minakawa; Atsushi Kasamatsu; Hirofumi Koike; Morihiro Higo; Dai Nakashima; Yukinao Kouzu; Yosuke Sakamoto; Katsunori Ogawara; Masashi Shiiba; Hideki Tanzawa; Katsuhiro Uzawa

Background Kinesin family member 4A (KIF4A), a microtubule-based motor protein, was implicated in regulation of chromosomal structure and kinetochore microtubule dynamics. Considering the functions of KIF4A, we assumed that KIF4A is involved in progression of oral squamous cell carcinomas (OSCCs) via activation of the spindle assembly checkpoint (SAC). However, little is known about the relevance of KIF4A in the behavior of OSCC. We investigated the KIF4A expression status and its functional mechanisms in OSCC. Methods The KIF4A expression levels in seven OSCC-derived cells were analyzed by quantitative reverse transcriptase-polymerase chain reaction and immunoblotting analyses. Using a KIF4A knockdown model, we assessed the expression of (SAC)-related molecules (BUB1, MAD2, CDC20, and cyclin B1), cell-cycle, and cellular proliferation. In addition to in vitro data, the clinical correlation between the KIF4A expression levels in primary OSCCs (n = 106 patients) and the clinicopathologic status by immunohistochemistry (IHC) also were evaluated. Results KIF4A mRNA and protein were up-regulated significantly (P < 0.05) in seven OSCC-derived cells compared with human normal oral keratinocytes. In the KIF4A knockdown cells, SAC activation was observed via increased BUB1 expression on the kinetochores, appropriate kinetochore localization of MAD2, down-regulation of CDC20, up-regulation of cyclin B1, and cell-cycle arrested at G2/M phase. The results showed that cellular proliferation of KIF4A knockdown cells decreased significantly (P < 0.05) compared with control cells. IHC showed that KIF4A expression in primary OSCCs was significantly (P < 0.05) greater than in the normal oral counterparts and that KIF4A-positive OSCCs were correlated closely (P < 0.05) with tumoral size. Conclusions Our results proposed for the first time that KIF4A controls cellular proliferation via SAC activation. Therefore, KIF4A might be a key regulator for tumoral progression in OSCCs.


PLOS ONE | 2015

Semaphorin7A Promotion of Tumoral Growth and Metastasis in Human Oral Cancer by Regulation of G1 Cell Cycle and Matrix Metalloproteases: Possible Contribution to Tumoral Angiogenesis.

Tomoaki Saito; Atsushi Kasamatsu; Katsunori Ogawara; Isao Miyamoto; Kengo Saito; Manabu Iyoda; Takane Suzuki; Yosuke Endo-Sakamoto; Masashi Shiiba; Hideki Tanzawa; Katsuhiro Uzawa

Background Semaphorins (SEMAs) consist of a large family of secreted and membrane-anchored proteins that are important in neuronal pathfinding and axon guidance in selected areas of the developing nervous system. Of them, SEMA7A has been reported to have a chemotactic activity in neurogenesis and to be an immunomodulator; however, little is known about the relevance of SEMA7A in the behaviors of oral squamous cell carcinoma (OSCC). Methods We evaluated SEMA7A expression in OSCC-derived cell lines and primary OSCC samples using quantitative reverse transcriptase-polymerase chain reaction, immunoblotting, and semiquantitative immunohistochemistry (sq-IHC). In addition, SEMA7A knockdown cells (shSEMA7A cells) were used for functional experiments, including cellular proliferation, invasiveness, and migration assays. We also analyzed the clinical correlation between SEMA7A status and clinical behaviors in patients with OSCC. Results SEMA7A mRNA and protein were up-regulated significantly (P<0.05) in OSCC-derived cell lines compared with human normal oral keratinocytes. The shSEMA7A cells showed decreased cellular growth by cell-cycle arrest at the G1 phase, resulting from up-regulation of cyclin-dependent kinase inhibitors (p21Cip1 and p27Kip1) and down-regulation of cyclins (cyclin D1, cyclin E) and cyclin-dependent kinases (CDK2, CDK4, and CDK6); and decreased invasiveness and migration activities by reduced secretion of matrix metalloproteases (MMPs) (MMP-2, proMMP-2, pro-MMP-9), and expression of membrane type 1- MMP (MT1-MMP). We also found inactivation of the extracellular regulated kinase 1/2 and AKT pathways, an upstream molecule of cell-cycle arrest at the G1 phase, and reduced secretion of MMPs in shSEMA7A cells. sq-IHC showed that SEMA7A expression in the primary OSCCs was significantly (P = 0.001) greater than that in normal counterparts and was correlated with primary tumoral size (P = 0.0254) and regional lymph node metastasis (P = 0.0002). Conclusion Our data provide evidence for an essential role of SEMA7A in tumoral growth and metastasis in OSCC and indicated that SEMA7A may play a potential diagnostic/therapeutic target for use in patients with OSCC.


BMC Cancer | 2015

Adenosine A2b receptor promotes progression of human oral cancer

Hiroki Kasama; Yosuke Sakamoto; Atsushi Kasamatsu; Atsushi Okamoto; Tomoyoshi Koyama; Yasuyuki Minakawa; Katsunori Ogawara; Hidetaka Yokoe; Masashi Shiiba; Hideki Tanzawa; Katsuhiro Uzawa

BackgroundAdenosine A2b receptor (ADORA2B) encodes an adenosine receptor that is a member of the G protein-coupled receptor superfamily. This integral membrane protein stimulates adenylate cyclase activity in the presence of adenosine. Little is known about the relevance of ADORA2B to human malignancy including oral squamous cell carcinoma (OSCC). We aimed to characterize the expression state and function of ADORA2B in OSCC.MethodsThe ADORA2B expression levels in nine OSCC-derived cells were analyzed by quantitative reverse transcriptase-polymerase chain reaction and immunoblotting analyses. Using an ADORA2B knockdown model, we assessed cellular proliferation and expression of hypoxia-inducible factor1α (HIF-1α). We examined the adenosine receptor expression profile under both normoxic and hypoxic conditions in the OSCC-derived cells. In addition to in vitro data, the clinical correlation between the ADORA2B expression levels in primary OSCCs (nu2009=u2009100 patients) and the clinicopathological status by immunohistochemistry (IHC) also was evaluated.ResultsADORA2B mRNA and protein were up-regulated significantly (pu2009<u20090.05) in seven OSCC-derived cells compared with human normal oral keratinocytes. Suppression of ADORA2B expression with shRNA significantly (pu2009<u20090.05) inhibited cellular proliferation compared with the control cells. HIF-1α also was down-regulated in ADORA2B knockdown OSCC cells. During hypoxia, ADORA2B expression was induced significantly (pu2009<u20090.05) in the mRNA and protein after 24xa0hours of incubation in OSCC-derived cells. IHC showed that ADORA2B expression in primary OSCCs was significantly (pu2009<u20090.05) greater than in the normal oral counterparts and that ADORA2B-positive OSCCs were correlated closely (pu2009<u20090.05) with tumoral size.ConclusionOur results suggested that ADORA2B controls cellular proliferation via HIF-1α activation, indicating that ADORA2B may be a key regulator of tumoral progression in OSCCs.


Molecular Carcinogenesis | 2015

Persephin: A potential key component in human oral cancer progression through the RET receptor tyrosine kinase-mitogen-activated protein kinase signaling pathway.

Takao Baba; Yosuke Sakamoto; Atsushi Kasamatsu; Yasuyuki Minakawa; Satoshi Yokota; Morihiro Higo; Hidetaka Yokoe; Katsunori Ogawara; Masashi Shiiba; Hideki Tanzawa; Katsuhiro Uzawa

Persephin (PSPN) is a neurotrophic factor of the glial cell line‐derived neurotrophic factor (GDNF) family that promotes survival of multiple populations of neurons. Little is known about the relevance of PSPN in human malignancy including oral squamous cell carcinoma (OSCC). This study was undertaken to evaluate PSPN mRNA and protein expression by analyzing cellular proliferation and the cell cycle in PSPN knockdown cells in vitro. PSPN mRNA and protein were significantly (Pu2009<u20090.05) up‐regulated in OSCC‐derived cells compared with human normal oral keratinocytes (nu2009=u20097). Cellular proliferation decreased significantly (Pu2009<u20090.05) in PSPN knockdown cells with reduced receptor tyrosine kinase (RTK) signaling, and cell‐cycle arrest at the G1 phase resulted from up‐regulation of the cyclin‐dependent kinase inhibitors (p21Cip1, p27Kip1, p15INK4B, and p16INK4A). Furthermore, the PSPN protein expression in 101 primary OSCCs was significantly (Pu2009<u20090.05) higher than in normal counterparts. Among the clinical variables analyzed, overexpression of PSPN also was related closely (Pu2009<u20090.05) to tumoral size. Our results suggested that PSPN is a possible key regulator of OSCC progression via PSPN‐RET‐mitogen‐activated protein kinase activation and that PSPN overexpression may have diagnostic potential for OSCC.


International Journal of Oncology | 2013

Lipocalin-2 is associated with radioresistance in oral cancer and lung cancer cells.

Masashi Shiiba; Kengo Saito; Kazuaki Fushimi; Takashi Ishigami; Keiji Shinozuka; Dai Nakashima; Yukinao Kouzu; Hirofumi Koike; Atsushi Kasamatsu; Yosuke Sakamoto; Katsunori Ogawara; Katsuhiro Uzawa; Yuichi Takiguchi; Hideki Tanzawa

The aim of the present study was to identify a target molecule that could predict the efficacy of radiotherapy in oral squamous cell carcinoma (OSCC). We used DNA microarray analysis to identify differences in gene expression after X-ray irradiation. We compared the gene expression profiles between X-ray (8 Gy)-irradiated Ca9-22 cells (an OSCC-derived cell line) and unirradiated Ca9-22 cells. A total of 167 genes with a 2-fold higher level of expression induced by X-ray irradiation were identified. Lipocalin-2 (LCN2) had the greatest increase in expression after X-ray irradiation, and it was categorized in a network that has cancer-related functions with the Ingenuity Pathway Analysis tool. Upregulated expression of LCN2 mRNA was validated by real-time quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) analysis. When the LCN2 gene was knocked down in OSCC cells (Ca9-22 and HSC-2) and lung cancer cells (A549) by using small interfering RNA, the radiosensitivity of these cells was enhanced. Our findings suggest that the overexpression of LCN2 is likely associated with radioresistance in oral cancer and lung cancer cells, and that LCN2 expression levels could be used to predict radioresistance. Thus, regulating the expression or function of LCN2 could enhance the radiation response, resulting in a favorable outcome of radiotherapy.


International Journal of Oncology | 2013

Protein O-fucosyltransferase 1: A potential diagnostic marker and therapeutic target for human oral cancer

Satoshi Yokota; Katsunori Ogawara; Ryota Kimura; Fumie Shimizu; Takao Baba; Yasuyuki Minakawa; Morihiro Higo; Atsushi Kasamatsu; Yosuke Endo-Sakamoto; Masashi Shiiba; Hideki Tanzawa; Katsuhiro Uzawa

Protein O-fucosyltransferase 1 (POFUT1) is the enzyme that adds O-fucose through O-glycosidic linkage to conserved serine or threonine residues in the epidermal growth factor-like repeats of a number of cellular surface and secreted proteins. Our previous study using microarray technology showed that significant upregulation of POFUT1 occurs in oral squamous cell carcinoma (OSCC)-derived cell lines compared to human normal oral keratinocytes. The aim of the present study was to examine the status of POFUT1 mRNA and protein expression in OSCC-derived cell lines and human primary OSCCs. POFUT1 mRNA was upregulated significantly (P<0.05 for both comparisons) in five OSCC-derived cell lines and primary OSCCs using quantitative reverse transcriptase-polymerase chain reaction. Immunohistochemistry data indicated that POFUT1 protein expression levels were consistent with mRNA expression status in OSCC-derived cell lines and primary OSCCs. Furthermore, POFUT1 expression status was correlated significantly (P=0.048) with the primary tumor size. The proliferation of POFUT1 knockdown cells was inhibited significantly compared with that of control cells. These results indicated that POFUT1 expression can contribute to cancer progression and that POFUT1 may serve as a diagnostic marker and a therapeutic target for OSCCs.


Cancer Medicine | 2013

Targeting phosphodiesterase 3B enhances cisplatin sensitivity in human cancer cells

Katsuhiro Uzawa; Atsushi Kasamatsu; Takao Baba; Katsuya Usukura; Yasuhiro Saito; Kentaro Sakuma; Manabu Iyoda; Yosuke Sakamoto; Katsunori Ogawara; Masashi Shiiba; Hideki Tanzawa

We previously reported that human squamous cell carcinoma (SCC) cell lines refractory to cis‐diaminedichloro‐platinum II (cisplatin [CDDP]) had significant upregulation of the phosphodiesterase 3B gene (PDE3B), suggesting that inhibiting PDE3B suppresses CDDP resistance. shRNA‐mediated PDE3B depletion in CDDP‐resistant cells derived from SCC cells and Hela cells and induced CDDP sensitivity and inhibited tumor growth with elevated cyclic GMP induction resulting in upregulation of the multidrug‐resistant molecule, but this did not occur in the 5‐fluorouracil‐resistant hepatocellular carcinoma cell lines. Furthermore, the antitumor growth effect of the combination of a PDE3B inhibitor (cilostazol) and CDDP in vivo was also greater than with either cilostazol or CDDP alone, with a significant increase in the number of apoptotic and cell growth‐suppressive cancer cells in CDDP‐resistance cell lines. Our results provided novel information on which to base further mechanistic studies of CDDP sensitization by inhibiting PDE3B in human cancer cells and for developing strategies to improve outcomes with concurrent chemotherapy.


Cancer Medicine | 2015

ANGPTL3 is a novel biomarker as it activates ERK/MAPK pathway in oral cancer

Tomoyoshi Koyama; Katsunori Ogawara; Atsushi Kasamatsu; Atsushi Okamoto; Hiroki Kasama; Yasuyuki Minakawa; Ken Shimada; Hidetaka Yokoe; Masashi Shiiba; Hideki Tanzawa; Katsuhiro Uzawa

Angiopoietin‐like 3 (ANGPTL3), which is involved in new blood vessel growth and stimulation of mitogen‐activated protein kinase (MAPK), is expressed aberrantly in several types of human cancers. However, little is known about the relevance of ANGPTL3 in the behavior of oral squamous cell carcinoma (OSCC). In this study, we evaluated ANGPTL3 mRNA and protein in OSCC‐derived cell lines (n = 8) and primary OSCCs (n = 109) and assessed the effect of ANGPTL3 on the biology and function of OSCCs in vitro and in vivo. Significant (P < 0.05) ANGPTL3 upregulation was detected in the cell lines and most primary OSCCs (60%) compared with the normal counterparts. The ANGPTL3 expression level was correlated closely (P < 0.05) with tumoral size. In patients with T3/T4 tumors, the overall survival rate with an ANGPTL3‐positive tumor was significantly (P < 0.05) lower than that of ANGPTL3‐negative cases. In vitro, cellular growth in ANGPTL3 knockdown cells significantly (P < 0.05) decreased with inactivated extracellular regulated kinase (ERK) and cell‐cycle arrest at the G1 phase resulting from upregulation of the cyclin‐dependent kinase inhibitors, including p21Cip1 and p27Kip1. We also observed a marked (P < 0.05) reduction in the growth in ANGPTL3 knockdown‐cell xenografts with decreased levels of phosphorylated ERK relative to control‐cell xenografts. The current data indicated that ANGPTL3 may play a role in OSCCs via MAPK signaling cascades, making it a potentially useful diagnostic/therapeutic target for use in patients with OSCC.

Collaboration


Dive into the Katsuhiro Uzawa'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

Hidetaka Yokoe

National Defense Medical College

View shared research outputs
Researchain Logo
Decentralizing Knowledge