Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Biochem Biophys Res Commun ; 457(1): 71-6, 2015 Jan 30.
Article in English | MEDLINE | ID: mdl-25550184

ABSTRACT

We reported previously that decorin (DCN) is significantly up-regulated in chemoresistant cancer cell lines. DCN is a small leucine-rich proteoglycan that exists and functions in stromal and epithelial cells. Accumulating evidence suggests that DCN affects the biology of several types of cancer by directly/indirectly targeting the signaling molecules involved in cell growth, survival, metastasis, and angiogenesis, however, the molecular mechanisms of DCN in chemoresistance and its clinical relevance are still unknown. Here we assumed that DCN silencing cells increase chemosusceptibility to S-1, consisted of tegafur, prodrug of 5-fluorouracil. We first established DCN knockdown transfectants derived from oral cancer cells for following experiments including chemosusceptibility assay to S-1. In addition to the in vitro data, DCN knockdown zenografting tumors in nude mice demonstrate decreasing cell proliferation and increasing apoptosis with dephosphorylation of AKT after S-1 chemotherapy. We also investigated whether DCN expression predicts the clinical responses of neoadjuvant chemotherapy (NAC) using S-1 (S-1 NAC) for oral cancer patients. Immunohistochemistry data in the preoperative biopsy samples was analyzed to determine the cut-off point for status of DCN expression by receiver operating curve analysis. Interestingly, low DCN expression was observed in five (83%) of six cases with complete responses to S-1 NAC, and in one (10%) case of 10 cases with stable/progressive disease, indicating that S-1 chemosensitivity is dramatically effective in oral cancer patients with low DCN expression compared with high DCN expression. Our findings suggest that DCN is a key regulator for chemoresistant mechanisms, and is a predictive immunomarker of the response to S-1 NAC and patient prognosis.


Subject(s)
Biomarkers, Tumor/metabolism , Decorin/metabolism , Mouth Neoplasms/drug therapy , Mouth Neoplasms/metabolism , Neoadjuvant Therapy , Oxonic Acid/therapeutic use , Tegafur/therapeutic use , Aged , Aged, 80 and over , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biopsy , Cell Line, Tumor , Drug Combinations , Female , Gene Knockdown Techniques , Humans , Immunoblotting , Immunohistochemistry , Male , Mice, Nude , Middle Aged , Mouth Neoplasms/pathology , Neoplasms, Squamous Cell/drug therapy , Neoplasms, Squamous Cell/metabolism , Neoplasms, Squamous Cell/pathology , Oxonic Acid/pharmacology , Tegafur/pharmacology , Xenograft Model Antitumor Assays
2.
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
3.
Oral Maxillofac Surg ; 17(1): 63-6, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22527656

ABSTRACT

BACKGROUND: Myoepithelioma, a generally benign tumor comprised of myoepithelial cells, is an uncommon salivary gland tumor. Among four morphologic variants of myoepithelioma, epithelioid type has not been reported in the oral and maxillofacial region. CASE REPORT: A 61-year-old man first noticed the mass 3 years previously. The oral examination revealed a firm, non-tender, and well-circumscribed mass in the middle of the hard palate. A magnetic resonance imaging scan showed a well-circumscribed mass with low signal intensity (T(1)-weighted image) or increased signal intensity (T(2)-weighted image). DISCUSSION: Immunohistochemically, the tumor cells in the present case reacted to the epithelial (CK HMW and CAM5.2) and the mesenchymal (vimentin) markers. However, myoepithelial markers (S-100 protein, α-smooth muscle actin, glial fibrillary acidic protein, and calponin), except p63, were not expressed in the tumor cells. These results indicated that the epithelial myoepithelioma cells differentiated into epithelial cells rather than myoepithelial cells. We believe that epithelioid myoepithelioma of the palate is a distinctive subtype of myoepithelioma that should be included in the differential diagnosis of tumors of the palate.


Subject(s)
Myoepithelioma/diagnosis , Palatal Neoplasms/diagnosis , Biomarkers, Tumor/analysis , Biopsy , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Myoepithelioma/pathology , Palatal Neoplasms/pathology , Palate, Hard/pathology
4.
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
5.
PLoS One ; 7(9): e45510, 2012.
Article in English | MEDLINE | ID: mdl-23029062

ABSTRACT

BACKGROUND: Annexins are calcium and phospholipid binding proteins that form an evolutionary conserved multigene family. Considerable evidence indicates that annexin A10 (ANXA10) is involved in tumoral progression, although little is known about its role in human oral carcinogenesis. In this study, we investigated the involvement of ANXA10 in oral squamous cell carcinoma (OSCC). METHODOLOGY/PRINCIPAL FINDINGS: ANXA10 mRNA and protein expressions were assessed by quantitative reverse transcriptase polymerase chain reaction and immunoblotting, and we conducted a proliferation assay and cell-cycle analysis in ANXA10 knockdown cells in vitro. We evaluated the correlation between the ANXA10 expression status in 100 primary OSCCs and the clinicopathological features by immunohistochemistry. ANXA10 mRNA and protein expression levels were up-regulated in all cellular lines examined (n = 7, p<0.05). ANXA10 knockdown cells showed that cellular proliferation decreased by inactivation of extracellular regulated kinase (ERK) (p<0.05), and cell-cycle arrest at the G1 phase resulted from up-regulation of cyclin-dependent kinase inhibitors. ANXA10 protein expression in primary OSCCs was also significantly greater than in normal counterparts (p<0.05), and higher expression was correlated with tumoral size (p = 0.027). CONCLUSIONS/SIGNIFICANCE: Our results proposed for the first time that ANXA10 is an indicator of cellular proliferation in OSCCs. Our results suggested that ANXA10 expression might indicate cellular proliferation and ANXA10 might be a potential therapeutic target for the development of new treatments for OSCCs.


Subject(s)
Annexins/genetics , Carcinoma, Squamous Cell/genetics , MAP Kinase Signaling System , Mouth Neoplasms/genetics , Aged , Aged, 80 and over , Annexins/metabolism , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Cycle/genetics , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Male , Middle Aged , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Neoplasm Staging , RNA Interference
6.
Int J Oncol ; 40(1): 47-52, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21879256

ABSTRACT

Heat shock factor 1 (HSF1) is responsible for expres-- sion of a large class of heat shock proteins that have been implicated in the malignant phenotype of human cancers. Little is known about the effect of a high level of HSF1 on the behavior of oral squamous cell carcinoma (OSCC). In this study, we assessed the value of HSF1 for predicting clinical outcomes in OSCC. Quantitative reverse transcriptase-polymerase chain reaction and Western blotting showed that the expressions of HSF1 mRNA and protein in OSCC-derived cell lines (HSC-2, HSC-3, HSC-4, Sa3, Ca9-22, KON and Ho-1-u-1) were elevated compared with those in human normal oral keratinocytes (P<0.05). Similar to in vitro data, HSF1 mRNA expression in primary OSCCs (n=50) was significantly greater than in normal counterparts (P<0.05). Since HSF1 was observed in the nucleus and cytoplasm by immu-- nohistochemistry, we investigated the correlation between the HSF1 expression status at each subcellular location and the clinical behavior of OSCCs. Among the clinical classifications, higher nuclear HSF1 expression was closely related to tumor size and histopathologic types (P<0.05). These results showed for the first time that nuclear HSF1 expression may contribute to cancer progression and that HSF1 might be a potential diagnostic biomarker and a therapeutic target for OSCCs.


Subject(s)
Biomarkers, Tumor/biosynthesis , Carcinoma, Squamous Cell/metabolism , DNA-Binding Proteins/biosynthesis , Mouth Neoplasms/metabolism , Transcription Factors/biosynthesis , Biomarkers, Tumor/genetics , Blotting, Western , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , DNA-Binding Proteins/genetics , Heat Shock Transcription Factors , Humans , Immunohistochemistry , Keratinocytes/metabolism , Mouth Neoplasms/diagnosis , Mouth Neoplasms/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...