Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Lab Invest ; 98(8): 980-988, 2018 08.
Article in English | MEDLINE | ID: mdl-29855544

ABSTRACT

Deoxynucleotidyl transferase terminal interacting protein 1 (DNTTIP1) forms a complex with histone deacetylase (HDAC); however, the relevance of DNTTIP1 in cancer remains unknown. The aim of this study was to examine DNTTIP1 expression and its functional mechanisms in oral squamous cell carcinomas (OSCCs). DNTTIP1 expression was analyzed by quantitative reverse transcriptase-polymerase chain reaction, immunoblotting analysis, and immunohistochemistry. The expression of DNTTIP1 was upregulated significantly in vitro and in vivo, and in patients with OSCC in whom DNTTIP1 was overexpressed and the expression level was correlated significantly (P < 0.05) with tumoral growth. DNTTIP1 knockdown (siDNTTIP1) cells showed depressed cellular proliferation by cell-cycle arrest at the G1 phase with high acetylation of p53 and upregulation of p21Cip1. Moreover, resveratrol, a HDAC inhibitor, controlled not only acetylated p53 status but also DNTTIP1 expression, leading to a similar phenotype of siDNTTIP1 cells. A marked (P < 0.05) reduction of tumoral growth in mouse xenograft models was observed with lower DNTTIP1 expression under the presence of this chemical reagent. Taken together, our results suggested that DNTTIP1-HDAC interaction promotes tumoral growth through deacetylation of p53 and that DNTTIP1 might be a critical therapeutic target in OSCCs.


Subject(s)
Carcinoma, Squamous Cell/genetics , Carrier Proteins/genetics , Cell Proliferation/genetics , Mouth Neoplasms/genetics , Nuclear Proteins/genetics , Aged , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Carrier Proteins/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , DNA-Binding Proteins , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice, Inbred BALB C , Mice, Nude , Mouth Neoplasms/drug therapy , Mouth Neoplasms/metabolism , Nuclear Proteins/metabolism , RNA Interference , Resveratrol/pharmacology , Transcription Factors , Xenograft Model Antitumor Assays/methods
2.
Mol Clin Oncol ; 2(4): 609-617, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24940505

ABSTRACT

The objective of this study was to elucidate the clinical characteristics of uncommon head and neck malignancies, such as non-squamous cell carcinoma (SCC), in order to improve patient outcomes. A total of 463 head and neck malignancies were retrospectively analyzed, with 43 cases (9.3%) diagnosed as non-SCC. The overall survival rate of patients with adenoid cystic carcinoma was significantly worse compared to that of patients with SCC. The 5-year survival rates were <50% for patients with malignant melanoma, adenocarcinoma, small-cell carcinoma and sarcomas. Distant metastasis to the lung was frequently observed in cases with a poor outcome. Non-SCC malignancies treated without surgery were associated with a worse outcome. Some non-SCC patients had a poor prognosis and distant metastasis was associated with an unsatisfactory outcome. Timely treatment and control of distant metastasis are essential and surgical treatment should be prioritized in non-SCC cases to improve patient outcomes.

3.
Mol Carcinog ; 53(7): 557-65, 2014 Jul.
Article in English | MEDLINE | ID: mdl-23371469

ABSTRACT

The human kallikrein-related peptidase family is comprised of 15 serine protease genes on chromosome 19q13.4. Our previous microarray analyses showed that the gene kallikrein-related peptidase 13 (KLK13) was down-regulated in oral squamous cell carcinoma (OSCC) cell lines. We evaluated the expression status of KLK13 in primary OSCCs and performed functional molecular experiments in OSCC cell lines. In 102 primary tumors studied, KLK13 expression significantly (P < 0.05) decreased compared with matched normal counterparts. Interestingly, KLK13-negative cases correlated significantly (P < 0.05) with regional lymph node metastasis. In vitro, cells overexpressing KLK13 (oeKLK13) had decreased invasiveness and motility and up-regulation of adhesion molecules (E-cadherin, α-catenin, ß-catenin, junction plakoglobin, plakophilin4, desmocollin2, desmoglein3, and desmoplakin) compared with control cells. A rescue experiment that transfected oeKLK13 cells with siRNA against KLK13 restored invasiveness and migration activities with down-regulated adhesion molecules. Based on our results, we concluded that KLK13 may play an important role in regulating cellular migration and invasiveness, making the loss of KLK13 a potential biomarker for early detection of lymph node metastasis in OSCCs.


Subject(s)
Biomarkers, Tumor/biosynthesis , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/pathology , Cell Adhesion Molecules/biosynthesis , Kallikreins/biosynthesis , Mouth Neoplasms/enzymology , Mouth Neoplasms/pathology , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Down-Regulation , Humans , Kallikreins/genetics , Lymphatic Metastasis , Neoplasm Invasiveness , RNA Interference , RNA, Small Interfering/genetics
4.
BMC Cancer ; 13: 555, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24261884

ABSTRACT

BACKGROUND: Glutamate decarboxylase 1 (GAD1), a rate-limiting enzyme in the production of γ-aminobutyric acid (GABA), is found in the GABAergic neurons of the central nervous system. Little is known about the relevance of GAD1 to oral squamous cell carcinoma (OSCC). We investigated the expression status of GAD1 and its functional mechanisms in OSCCs. METHODS: We evaluated GAD1 mRNA and protein expressions in OSCC-derived cells using real-time quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) and immunoblotting analyses. To assess the critical functions of GAD1, i.e., cellular proliferation, invasiveness, and migration, OSCC-derived cells were treated with the shRNA and specific GAD1 inhibitor, 3-mercaptopropionic acid (3-MPA). GAD1 expression in 80 patients with primary OSCCs was analyzed and compared to the clinicopathological behaviors of OSCC. RESULTS: qRT-PCR and immunoblotting analyses detected frequent up-regulation of GAD1 in OSCC-derived cells compared to human normal oral keratinocytes. Suppression of nuclear localization of ß-catenin and MMP7 secretion was observed in GAD1 knockdown and 3-MPA-treated cells. We also found low cellular invasiveness and migratory abilities in GAD1 knockdown and 3-MPA-treated cells. In the clinical samples, GAD1 expression in the primary OSCCs was significantly (P < 0.05) higher than in normal counterparts and was correlated significantly (P < 0.05) with regional lymph node metastasis. CONCLUSIONS: Our data showed that up-regulation of GAD1 was a characteristic event in OSCCs and that GAD1 was correlated with cellular invasiveness and migration by regulating ß-catenin translocation and MMP7 activation. GAD1 might play an important role in controlling tumoral invasiveness and metastasis in oral cancer.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Glutamate Decarboxylase/physiology , Matrix Metalloproteinase 7/metabolism , Mouth Neoplasms/enzymology , beta Catenin/metabolism , 3-Mercaptopropionic Acid/pharmacology , Adult , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/secondary , Cell Line, Tumor , Cell Movement , Cell Nucleus/metabolism , Cell Proliferation , Enzyme Activation , Glutamate Decarboxylase/antagonists & inhibitors , Humans , Middle Aged , Mouth Neoplasms/pathology , Neoplasm Invasiveness , Protein Transport
5.
Int J Oncol ; 42(4): 1197-204, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23403985

ABSTRACT

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.


Subject(s)
Acute-Phase Proteins/metabolism , Lipocalins/metabolism , Proto-Oncogene Proteins/metabolism , Radiation Tolerance , Acute-Phase Proteins/genetics , Cell Line, Tumor , Cell Survival/radiation effects , Humans , Lipocalin-2 , Lipocalins/genetics , Lung Neoplasms , Mouth Neoplasms , Oligonucleotide Array Sequence Analysis , Protein Interaction Maps , Proto-Oncogene Proteins/genetics , Transcriptome/radiation effects
6.
J Cancer Res Clin Oncol ; 139(1): 123-30, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22986808

ABSTRACT

PURPOSE: Tripeptidyl peptidase II (TPP2), a member of the family of eukaryotic serine peptidase, has been implicated in DNA repair, cellular division, and apoptosis. The aim of this study was to examine TPP2 expression and its functional mechanisms in oral squamous cell carcinoma (OSCC). METHODS: TPP2 mRNA and protein expression in seven OSCC-derived cells (Ca9-22, HSC-2, HSC-3, HSC-4, HO-1-N-1, H1, and Sa3) was analyzed by quantitative reverse transcriptase-polymerase chain reaction and immunoblotting analyses. Since previous studies indicated that TPP2 might control chromosomal division, we investigated cellular proliferation and spindle assembly checkpoint (SAC) molecules, MAD2 and CCNB1. In addition, we evaluated the correlation between TPP2 expression levels in primary OSCCs (n = 108 specimens) and the clinicopathologic status by immunohistochemistry (IHC). RESULTS: TPP2 mRNA and protein were significantly (P < 0.05) up-regulated in OSCC-derived cells compared with human normal oral keratinocytes. Suppression of TPP2 expression with shRNA significantly (P < 0.05) inhibited cellular proliferation compared with the control cells. In addition, appropriate localization of MAD2 and up-regulation of CCNB1 were observed in TPP2 knockdown OSCC cells. IHC showed that TPP2 expression in primary OSCCs was significantly (P < 0.001) greater than that in the normal oral counterparts, and the TPP2-positive cases were significantly (P < 0.05) correlated with tumor size. CONCLUSION: The current study showed that overexpression of TPP2 occurs frequently during oral carcinogenesis and might be associated with OSCC progression via SAC activation.


Subject(s)
Aminopeptidases/metabolism , Calcium-Binding Proteins/metabolism , Carcinoma, Squamous Cell/enzymology , Cell Cycle Proteins/metabolism , Cyclin B1/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , M Phase Cell Cycle Checkpoints , Mouth Neoplasms/enzymology , Repressor Proteins/metabolism , Serine Endopeptidases/metabolism , Aged , Aminopeptidases/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Down-Regulation/drug effects , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Immunoblotting , Japan , Lymphatic Metastasis , Mad2 Proteins , Male , Middle Aged , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Neoplasm Staging , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Serine Endopeptidases/genetics , Transfection , Up-Regulation
7.
Int J Oncol ; 42(1): 141-50, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23123766

ABSTRACT

The four and a half LIM domains 1 (FHL1) gene has been related to carcinogenesis. However, the expression status of FHL1 in human oral squamous cell carcinoma (OSCC) remains unclear and the detailed mechanism of gene silencing is poorly understood. The aim of this study was to examine the FHL1 expression level and its regulatory mechanism in OSCCs. Quantitative reverse-transcriptase-polymerase chain reaction (PCR) and western blotting showed significant downregulation of FHL1 in all OSCC-derived cell lines (Sa3, HSC-2, HSC-3, HSC-4, HO-1-u-1, HO-1-N-1, KON and Ca9-22) compared to human normal oral keratinocytes. We also found that FHL1 mRNA expression was frequently downregulated (P<0.01) in 51 (86.4%) of 59 primary OSCCs compared with the corresponding normal oral tissues, while there was no significant difference between the status of the FHL1 protein expression in OSCCs and the clinicopathological features. Using methylation-specific PCR, we detected methylated FHL1 in all cell lines and treatment with the DNA methyltransferase inhibitor, 5-aza-2'-deoxycytidine restored the FHL1 expression. However, no significant restoration of FHL1 expression was observed using sodium butyrate, an inhibitor of histone deacetylase and chromatin immunoprecipitation showed that histone H3 lysine 9 in the FHL1 promoter region was significantly acetylated. In addition, no mutation in the entire coding region of the FHL1 gene was found. Therefore, our data suggested that inactivation of the FHL1 gene is a frequent event during oral carcinogenesis and that the mechanism of FHL1 downregulation in OSCCs is through DNA methylation of the promoter region rather than histone deacetylation or mutation.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/genetics , DNA Methylation , Epigenomics , Intracellular Signaling Peptides and Proteins/genetics , LIM Domain Proteins/genetics , Mouth Neoplasms/genetics , Muscle Proteins/genetics , Aged , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/secondary , Case-Control Studies , Female , Gene Silencing , Humans , Lymphatic Metastasis , Male , Middle Aged , Mouth/metabolism , Mouth/pathology , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Neoplasm Staging , Prevalence , Prognosis , Promoter Regions, Genetic/genetics , Tumor Cells, Cultured
8.
J Cancer Res Clin Oncol ; 139(4): 533-42, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23207790

ABSTRACT

PURPOSE: Sec8, a component of the exocyst complex, has been implicated in tethering of secretory vesicles to specific regions on the plasma membrane. To investigate the involvement of Sec8 in oral squamous-cell carcinoma (OSCC), we evaluated the expression status and effect of Sec8 in OSCC cell lines. METHODS: Sec8 mRNA and protein expressions in human OSCC cell lines were assessed by quantitative reverse transcriptase-polymerase chain reaction and immunoblotting. Functional analyses, proliferation assay, invasiveness assay, and gelatin zymography in Sec8 knockdown cells were performed. Also the correlation between Sec8 expression and the clinicopathological features in 98 primary OSCCs samples was evaluated by immunohistochemistry. RESULTS: Sec8 mRNA and protein expression were significantly up-regulated in all cell lines (p < 0.05). Sec8 knockdown cells were characterized by reduced cellular proliferation, invasiveness, and secretion of matrix metalloproteinases (MMPs) (MMP-2, proMMP-2, and proMMP-9). Sec8 protein expression in primary OSCCs also was significantly (p < 0.05) greater than in normal counterparts, and higher Sec8 expression was correlated with tumor size (p = 0.03). CONCLUSIONS: Our results suggested for the first time that Sec8 might play a specific role in OSCC progression by mediating MMP secretion.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mouth Mucosa/metabolism , Mouth Neoplasms/metabolism , Vesicular Transport Proteins/metabolism , Adult , Aged , Aged, 80 and over , Apoptosis , Blotting, Western , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Adhesion , Cell Movement , Cell Proliferation , Disease Progression , Female , Humans , Immunoenzyme Techniques , Lymphatic Metastasis , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Middle Aged , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Vesicular Transport Proteins/antagonists & inhibitors , Vesicular Transport Proteins/genetics
9.
PLoS One ; 8(12): e85951, 2013.
Article in English | MEDLINE | ID: mdl-24386490

ABSTRACT

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.


Subject(s)
Carcinoma, Squamous Cell/metabolism , G2 Phase Cell Cycle Checkpoints , Gene Expression Regulation, Neoplastic , Kinesins/biosynthesis , Mouth Neoplasms/metabolism , Neoplasm Proteins/biosynthesis , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Female , Humans , Kinesins/genetics , Male , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Neoplasm Proteins/genetics , Up-Regulation/genetics
10.
Int J Oncol ; 40(5): 1414-22, 2012 May.
Article in English | MEDLINE | ID: mdl-22267082

ABSTRACT

ADAMs are a disintegrin and metalloproteinase family of membrane-associated metalloproteinases characterized by their multidomain structure, and have been reported to be associated with various malignant tumors. The aim of this study was to identify crucial members of the ADAM family in oral squamous cell carcinoma (OSCC), and to reveal their biological function and clinical significance. To clarify whether ADAM family genes are involved in OSCC, changes in the expression profile were investigated by real-time quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) analysis and immunohistochemical analysis. Functional analysis was performed by comparing cellular proliferation of siADAM-transfected cell lines and parental cell lines. Real-time qRT-PCR analysis identified significantly upregulated expression of ADAM12 in OSCC-derived cell lines. This was validated in OSCC samples using real-time qRT-PCR and immuno-histochemical staining. ADAM12 expression was correlated with TNM classification; significantly greater expression of ADAM12 was observed in tumors with higher T classification and more advanced stages. Moreover, siADAM12-transfected cells showed both a suppressed proliferation rate and increased transforming growth factor (TGF)-ß3 expression. Our data indicate that ADAM12 is overexpressed in OSCC and might accelerate cellular proliferation. Its function may be associated with TGF-ß signaling. This study suggests that controlling the expression or activity of ADAM12 could be a useful strategy in the development of an effective cure for OSCC.


Subject(s)
ADAM Proteins/metabolism , Carcinoma, Squamous Cell/enzymology , Membrane Proteins/metabolism , Mouth Neoplasms/enzymology , ADAM Proteins/genetics , ADAM12 Protein , Aged , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation , Chi-Square Distribution , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Male , Membrane Proteins/genetics , Middle Aged , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Neoplasm Staging , Prognosis , RNA Interference , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transfection , Transforming Growth Factor beta3/metabolism , Tumor Burden , Up-Regulation
11.
Int J Cancer ; 130(12): 2903-11, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-21796630

ABSTRACT

Dermatopontin (DPT), a component of the extracellular matrix (ECM), is involved in promotion of cellular adhesion and ECM assembly activities. However, the role of DPT in the pathogenesis of carcinoma is unclear. We evaluated DPT expression in human oral cancer and its possible roles including cellular adhesion and invasiveness. We first investigated the DPT mRNA and protein expression status in human oral squamous cell carcinoma (OSCC)-derived cells. Real-time quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) and immunoblotting analysis detected frequent downregulation of DPT in OSCC-derived cells compared to human normal oral keratinocytes. To assess the epigenetic regulation of DPT, OSCC-derived cells were treated with a histone deacetylase inhibitor, sodium butyrate (NaB). NaB restored the DPT expression in OSCC-derived cells. DPT-overexpressed cells were examined whether DPT could contribute to cellular adhesion and invasiveness. Markedly, increased adhesion and decreased invasiveness in DPT-overexpressed cells were found compared to mock-transfected cells. Adhesion of DPT-overexpressed cells was inhibited by α3ß1 integrin functional blocking antibody. OSCC-derived cells treated with NaB also decreased invasiveness. The expression status of DPT in primary OSCCs (n = 97) was analyzed and compared to clinicopathological behavior. DPT expression in primary OSCCs was significantly lower (p < 0.05) than in the normal counterparts and was correlated significantly (p < 0.05) with regional lymph node metastasis. Our data provided strong evidence that downregulation of DPT is a characteristic event in OSCCs and that DPT was correlated with cellular adhesion and invasiveness. Therefore, DPT might play an important role in regulating tumor invasion and metastasis.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , Extracellular Matrix Proteins/metabolism , Lymphatic Metastasis , Mouth Neoplasms/pathology , Aged , Aged, 80 and over , Antibodies, Blocking , Butyrates/pharmacology , Butyric Acid/pharmacology , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/secondary , Cell Adhesion , Cell Line, Tumor , Cell Proliferation , Chondroitin Sulfate Proteoglycans/biosynthesis , Extracellular Matrix Proteins/biosynthesis , Female , Gene Expression Regulation, Neoplastic , Histone Deacetylase Inhibitors/pharmacology , Humans , Integrin alpha3beta1/immunology , Keratinocytes/metabolism , Male , Middle Aged , Mouth Neoplasms/metabolism , Neoplasm Invasiveness , RNA, Messenger/biosynthesis
12.
Oncol Rep ; 24(1): 241-9, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20514468

ABSTRACT

Serpins (serine protease inhibitors) are known as a diverse family of protease inhibitors; however, various other biological activities including tumor suppression, have been recently reported for these molecules. To clarify whether members of the serpin family are involved in OSCC (oral squamous cell carcinoma), global gene screening using microarray analysis was performed with OSCC-derived cell lines. A trend toward diminished expression was shown for some SERPIN genes located on 11q12-q13.1 and 18q21. mRNA expression of SERPIN genes at these chromosome regions was therefore analyzed using real-time quantitative RT-PCR (qRT-PCR) in 55 OSCC samples and matched normal tissue. Statistically significant decreases in expression were found for SERPINB12 (P=0.001), SERPINB13 (P=0.001), SERPINB4 (P=0.042), SERPINB3 (P<0.001), SERPINB11 (P<0.001), SERPINB7 (P=0.021) and SERPINB2 (P=0.018). All of these genes are located on 18q21, the known location of the serpin gene cluster. The results strongly suggest that this chromosome region plays a crucial role in OSCC. Some serpin members in the region might be involved in tumor suppression, or there might be unidentified tumor suppressor genes within or near the chromosome region.


Subject(s)
Carcinoma, Squamous Cell/genetics , Chromosomes, Human, Pair 18 , Mouth Neoplasms/genetics , Serpins/genetics , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/pathology , Chromosome Mapping , Chromosomes, Human, Pair 18/genetics , Down-Regulation/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Male , Middle Aged , Mouth Neoplasms/pathology , Oligonucleotide Array Sequence Analysis , Tumor Cells, Cultured
13.
Int J Cancer ; 126(2): 437-49, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19569180

ABSTRACT

Resistance to cisplatin is a major obstacle to successful treatment of head and neck squamous cell carcinoma (HNSCC). To investigate the molecular mechanism of this resistance, we compared the gene expression profiles between the cisplatin-sensitive SCC cell lines (Sa-3, H-1 and KB) and the cisplatin-resistant cell lines established from them (Sa-3R, H-1R and KB-R) using Affymetrix U133 Plus 2.0 microarray. We identified 199 genes differentially expressed in each group. To identify important functional networks and ontologies to cisplatin resistance, the 199 genes were analyzed using the Ingenuity Pathway Analysis Tool. Fifty-one of these genes were mapped to genetic networks, and we validated the top-10 upregulated genes by real-time reverse transcriptase-polymerase chain reaction. Five novel genes, LUM, PDE3B, PDGF-C, NRG1 and PKD2, showed excellent concordance with the microarray data. In 48 patients with oral SCC (OSCC), positive immunohistochemical staining for the five genes correlated with chemoresistance to cisplatin-based combination chemotherapy. In addition, the expression of the five genes predicted the patient outcomes with chemotherapy. Furthermore, siRNA-directed suppressed expression of the five genes resulted in enhanced susceptibility to cisplatin-mediated apoptosis. These results suggested that these five novel genes have great potential for predicting the efficacy of cisplatin-based chemotherapy against OSCC. Global gene analysis of cisplatin-resistant cell lines may provide new insights into the mechanisms underlying clinical cisplatin resistance and improve the efficacy of chemotherapy for human HNSCC.


Subject(s)
Carcinoma, Squamous Cell/genetics , Cisplatin/pharmacology , Drug Resistance, Neoplasm/genetics , Head and Neck Neoplasms/genetics , Aged , Antineoplastic Agents/pharmacology , Blotting, Western , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Chondroitin Sulfate Proteoglycans/genetics , Chondroitin Sulfate Proteoglycans/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/genetics , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Genetic Predisposition to Disease/genetics , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Immunohistochemistry , Keratan Sulfate/genetics , Keratan Sulfate/metabolism , Lumican , Lymphokines/genetics , Lymphokines/metabolism , Male , Middle Aged , Neuregulin-1/genetics , Neuregulin-1/metabolism , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , TRPP Cation Channels/genetics , TRPP Cation Channels/metabolism
14.
J Cancer Res Clin Oncol ; 135(7): 943-9, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19101730

ABSTRACT

PURPOSE: To determine the potential involvement of ANXA1 in oral squamous-cell carcinoma (OSCC), we evaluated the ANXA1 protein expression in oral premalignant lesions (OPLs) and OSCCs and correlated the results with clinicopathologic variables. METHODS: Matched normal and tumour specimens of 44 primary OSCCs and 28 OPLs were analyzed for ANXA1 subcellular localization and protein expression level by immunohistochemistry (IHC). Correlations between ANXA1-IHC staining scores of OSCCs and clinicopathologic features were evaluated by Fisher's exact test. RESULTS: Markedly down-regulation of ANXA1 protein expression was identified on the plasma membrane of epithelial cells in OSCCs (P < 0.001) and OPLs (P = 0.001) compared with normal counterparts. Moreover, loss of plasma membranous ANXA1 expression was significantly correlated with the poorly differentiated status of OSCC cells (P = 0.012). CONCLUSIONS: Our findings suggest that loss of ANXA1 is frequent and early event during oral carcinogenesis and that ANXA1 could contribute to maintaining epithelial differentiation in OSCC.


Subject(s)
Annexin A1/metabolism , Carcinoma, Squamous Cell/metabolism , Cell Differentiation , Epithelial Cells/physiology , Mouth Neoplasms/metabolism , Precancerous Conditions/metabolism , Carcinoma, Squamous Cell/pathology , Cell Differentiation/physiology , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cell Nucleus/pathology , Disease Progression , Down-Regulation , Epithelial Cells/metabolism , Humans , Mouth/metabolism , Mouth/pathology , Mouth Mucosa/metabolism , Mouth Mucosa/pathology , Mouth Neoplasms/pathology , Neoplasm Staging
15.
Hum Pathol ; 40(1): 83-91, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18789482

ABSTRACT

Autophagy is a dynamic process of subcellular degradation, which has recently sparked great interest because it is involved in various developmental processes and various diseases including cancer. Autophagy-related 16-like 1 is a component of a large protein complex essential for autophagosome formation. We previously applied proteomic methods to characterize differentially expressed proteins in oral squamous cell carcinoma cells and detected significantly high expression levels of autophagy-related 16-like 1 in oral squamous cell carcinoma-derived cell lines compared to human normal oral keratinocytes. In the current study, to further determine the potential involvement of autophagy-related 16-like 1 in oral squamous cell carcinoma, we evaluated the state of autophagy-related 16-like 1 protein expression in human oral premalignant lesions and primary oral squamous cell carcinomas, and correlated the results with clinicopathologic variables. Autophagy-related 16-like 1 immunoreaction was predominant in a variety of subcellular components of oral squamous cell carcinoma tissues, including the cytoplasm and plasma membrane of malignant cells (45% and 39%, respectively) and peritumoral and intratumoral stroma (52%), whereas all of the components in normal tissues had no or faint autophagy-related 16-like 1 expression. In addition, high stromal expression of autophagy-related 16-like 1 was associated significantly with lymphovascular invasion of tumor cells (P = .037) and positive lymph node status (P = .015). Furthermore, cytoplasmic and plasma membranous autophagy-related 16-like 1 were also expressed in abundance in the oral premalignant lesion cells (74% and 32%, respectively). Our finding suggests that dysregulation of autophagy-related 16-like 1 protein expression is a frequent and early event during oral carcinogenesis and could affect the malignant behavior of oral squamous cell carcinoma cells.


Subject(s)
Autophagy , Carcinoma, Squamous Cell/metabolism , Lymph Nodes/metabolism , Mouth Neoplasms/metabolism , Neoplasm Proteins/metabolism , Adult , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/pathology , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cytoplasm/metabolism , Female , Humans , Immunohistochemistry , Lymph Nodes/pathology , Lymphatic Metastasis/pathology , Male , Middle Aged , Mouth Neoplasms/pathology , Transcription Factors
16.
BMC Cancer ; 8: 39, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18237446

ABSTRACT

BACKGROUND: Gelsolin-like actin-capping protein (CapG) is a ubiquitous gelsolin-family actin-modulating protein involved in cell signalling, receptor-mediated membrane ruffling, phagocytosis, and motility. CapG has generated great interest due to its oncogenic function in the control of cell migration or invasion in a variety of cancer cells. We previously applied proteomic methods to characterize differentially expressed proteins in oral squamous-cell carcinoma (OSCC) cells and detected significantly high expression levels of CapG in OSCC-derived cell lines compared to human normal oral keratinocytes. In the current study, to further determine the potential involvement of CapG in OSCC, we evaluated the status of CapG protein and mRNA expression in human oral premalignant lesions (OPLs) and primary OSCCs and correlated the results with clinicopathologic variables. METHODS: Matched normal and tumour tissue sections of 79 human primary OSCCs and 28 OPLs were analyzed for CapG expression by immunohistochemistry (IHC). Correlations between CapG-immunohistochemical staining scores of OSCCs and clinicopathologic features were evaluated by Fisher's exact test. Real-time quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) was used to estimate CapG expression at the mRNA level. RESULTS: In IHC, substantial up-regulation of CapG protein was observed in primary OSCCs (52%) and OPLs (64%), whereas corresponding normal tissues showed consistently weak or absent immunoreactivity of CapG. qRT-PCR data were consistent with the protein expression status. Moreover, CapG expression was correlated with the TNM stage grading of OSCCs. CONCLUSION: Our finding of frequent dysregulated expression of CapG in premalignant and malignant lesions together with an association with an advanced clinical disease stage suggests that CapG could contribute to cancer development and progression and that CapG may have potential as a biomarker and a therapeutic target for OSCC.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Gene Expression Regulation, Neoplastic/physiology , Microfilament Proteins/biosynthesis , Mouth Neoplasms/metabolism , Nuclear Proteins/biosynthesis , Precancerous Conditions/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Carrier Proteins/analysis , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cell Line, Tumor , Gelsolin/analysis , Gelsolin/biosynthesis , Gelsolin/genetics , Humans , Immunohistochemistry , Microfilament Proteins/analysis , Microfilament Proteins/genetics , Mouth/metabolism , Mouth/pathology , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Nuclear Proteins/analysis , Nuclear Proteins/genetics , Precancerous Conditions/genetics , Precancerous Conditions/pathology
17.
Int J Oncol ; 31(5): 1021-8, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17912427

ABSTRACT

Our previous study using proteomic profiling demonstrated significant up-regulation of Septin1, a conserved family of GTPase proteins, in oral squamous-cell carcinoma (OSCC)-derived cell lines. In the current study, to determine the potential involvement of Septin1 in oral carcinogenesis, we evaluated the state of septin1 protein/mRNA expression in OSCC-derived cell lines, oral premalignant lesions (OPLs), and primary OSCCs. A significant (P<0.05) increase in Septin1 expression was evident in all OSCC-derived cell lines examined compared to human normal oral keratinocytes (HNOKs) and OPLs. In immunohistochemistry, while the vast majority of the OSCCs (89%) were positive for Septin1, no immunoreaction was observed in corresponding normal tissues and OPLs. In addition, real-time quantitative reverse transcriptase-polymerase chain reaction (QRT-PCR) data were consistent with the protein expression status. These results suggest that Septin1 expression could contribute to cancer progression, proliferation, or both, and that Septin1 may be a potential diagnostic marker of highly active cancer and a therapeutic target for OSCCs.


Subject(s)
Carcinoma, Squamous Cell/etiology , Mouth Neoplasms/etiology , Phosphoric Monoester Hydrolases/physiology , Adult , Aged , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/mortality , Cell Line, Tumor , Female , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Male , Middle Aged , Mouth Neoplasms/metabolism , Mouth Neoplasms/mortality , Phosphoric Monoester Hydrolases/analysis , Phosphoric Monoester Hydrolases/genetics
18.
Oncol Rep ; 16(5): 1003-8, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17016584

ABSTRACT

The oncogenic transcription factor PAX5 is an important developmental regulator and is implicated in the pathogenesis of several malignancies. The PAX5 gene is involved in medulloblastoma, non-Hodgkin's lymphoma, transitional cell carcinoma of the bladder, neuroblastoma, breast cancer and SCC. In the current study, to determine the potential involvement of PAX5 in oral squamous-cell carcinoma (OSCC) and leukoplakias, we evaluated the status of PAX5 mRNA and protein expression in OSCC cell lines, human primary OSCCs, and leukoplakias by real-time quantitative reverse transcriptase-polymerase chain reaction, Western blot analysis, and immunohistochemistry. A significant increase in PAX5 expression was observed in all OSCC-derived cell lines examined compared to human normal oral keratinocytes (HNOKs). In immunohistochemistry, 78% of tumors and 42% of leukoplakias examined were positive for PAX5, while no immunoreaction was observed in corresponding normal tissues. The results suggest that PAX5 plays an important role during oral carcinogenesis, especially in the early stage, and that the gene may have potential as a biomarker and therapeutic target for OSCC.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Mouth Neoplasms/metabolism , PAX5 Transcription Factor/biosynthesis , Adult , Aged , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Female , Humans , Immunohistochemistry , Leukoplakia/genetics , Leukoplakia/metabolism , Male , Middle Aged , Mouth Neoplasms/genetics , PAX5 Transcription Factor/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
19.
Int J Radiat Oncol Biol Phys ; 65(3): 867-75, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16751068

ABSTRACT

PURPOSE: The purpose of this study was to assess the gene expression changes in oral squamous cell carcinoma (OSCC) cells after carbon ion irradiation. METHODS AND MATERIALS: Three OSCC cell lines (HSC2, Ca9-22, and HSC3) were irradiated with accelerated carbon ion beams or X-rays using three different doses. The cellular sensitivities were determined by clonogenic survival assay. To identify genes the expression of which is influenced by carbon ion irradiation in a dose-dependent manner, we performed Affymetrix GeneChip analysis with HG-U133 plus 2.0 arrays containing 54,675 probe sets. The identified genes were analyzed using the Ingenuity Pathway Analysis Tool to investigate the functional network and gene ontology. Changes in mRNA expression in the genes were assessed by real-time reverse transcriptase-polymerase chain reaction. RESULTS: We identified 98 genes with expression levels that were altered significantly at least twofold in each of the three carbon-irradiated OSCC cell lines at all dose points compared with nonirradiated control cells. Among these, SPHK1, the expression of which was significantly upregulated by carbon ion irradiation, was modulated little by X-rays. The function of SPHK1 related to cellular growth and proliferation had the highest p value (p = 9.25e-7 to 2.19e-2). Real-time reverse transcriptase-polymerase chain reaction analysis showed significantly elevated SPHK1 expression levels after carbon ion irradiation (p < 0.05), consistent with microarray data. Clonogenic survival assay indicated that carbon ion irradiation could induce cell death in Ca9-22 cells more effectively than X-rays. CONCLUSIONS: Our findings suggest that SPHK1 helps to elucidate the molecular mechanisms and processes underlying the biologic response to carbon ion beams in OSCC.


Subject(s)
Adaptor Proteins, Signal Transducing/radiation effects , Carbon/therapeutic use , Carcinoma, Squamous Cell , Heavy Ion Radiotherapy , Microarray Analysis/methods , Mouth Neoplasms , Adaptor Proteins, Signal Transducing/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/radiotherapy , Cell Line, Tumor , Cell Survival/genetics , Cell Survival/radiation effects , Gene Expression/radiation effects , Gene Expression Profiling/methods , Humans , Linear Energy Transfer , Mouth Neoplasms/genetics , Mouth Neoplasms/radiotherapy , Nucleic Acid Hybridization/methods , RNA, Neoplasm/genetics
20.
Int J Mol Med ; 17(4): 567-73, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16525711

ABSTRACT

This study was designed to disclose detailed genetic mechanisms in salivary gland tumors (SGTs) for development of novel independent marker. We constructed an in-house cDNA microarray carrying 2,201 cDNA clones derived from SGT and oral squamous cell carcinoma cDNA libraries. Four cell lines that originated from the SGT-derived cell lines were analyzed using this microarray system. The genes identified by our microarray system were further analyzed at the mRNA or protein expression level in other types of human cancer cell lines and clinical samples (ten normal salivary glands [NSGs], eleven pleomorphic adenomas, ten adenoid cystic carcinomas and three adenocarcinomas). Two up-regulated genes and six down-regulated genes were identified in common when compared with the control RNA. Of the up-regulated genes, WISP-2, which plays an important role in breast carcinogenesis, was selected for further analyses. We found a higher expression of the WISP-2 gene in the SGT-derived cell lines compared with other types of human cancer cell lines. Furthermore, WISP-2 mRNA and protein expression levels in NSGs were significantly higher than those in SGTs. These results suggest that WISP-2 could be a reliable independent marker and that down-regulation or loss of the WISP-2 gene may be associated with the development of SGTs.


Subject(s)
Gene Expression Regulation, Neoplastic , Intercellular Signaling Peptides and Proteins/genetics , Neoplasm Proteins/genetics , Salivary Gland Neoplasms/genetics , Transcription Factors/genetics , CCN Intercellular Signaling Proteins , Cell Line, Tumor , Gene Library , Humans , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/metabolism , Neoplasm Proteins/metabolism , Oligonucleotide Array Sequence Analysis/methods , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins , Reverse Transcriptase Polymerase Chain Reaction , Salivary Gland Neoplasms/pathology , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...