Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
Add more filters










Publication year range
1.
Asian Pac J Cancer Prev ; 19(3): 667-675, 2018 Mar 27.
Article in English | MEDLINE | ID: mdl-29580038

ABSTRACT

Background: Insulin-like growth factor-binding protein 7 (IGFBP7) has been found to be a tumor suppressor in several human cancers, but the role of IGFBP7 in gastric cancer has not yet been fully investigated. Herein, we examined the epigenetic downregulation of IGFBP7 expression in gastric cancer. Methods: Expression and methylation of IGFBP7 in gastric cancer cells and primary gastric cancer patients were determined using qRT-PCR, western blot, immunohistochemistry, and methylation specific-PCR, respectively. The effects of IGFBP7 on gastric cancer cells were investigated by various experimental conditions, such as proliferation, colony formation, apoptosis, invasion, and migration assay. Results: IGFBP7 methylation was inversely correlated with IGFBP7 expression in gastric cancer. Univariate and multivariate analysis showed that IGFBP7 expression and tumor stage were independent prognostic factors. IGFBP7 knockdown increased gastric cancer cell growth, invasion, and migration, whereas IGFBP7 overexpression in gastric cancer cells induced cell growth inhibition and apoptosis. Conclusion: Our data suggest that IGFBP7 functions as a tumor suppressor in gastric cancer via an epigenetic pathway.


Subject(s)
Adenocarcinoma/pathology , Apoptosis , Biomarkers, Tumor/metabolism , Epigenomics , Gene Expression Regulation, Neoplastic , Insulin-Like Growth Factor Binding Proteins/metabolism , Stomach Neoplasms/pathology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Biomarkers, Tumor/genetics , Cell Movement , Cell Proliferation , DNA Methylation , Female , Follow-Up Studies , Humans , Insulin-Like Growth Factor Binding Proteins/genetics , Male , Middle Aged , Prognosis , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Tumor Cells, Cultured
2.
Cancer Res Treat ; 50(1): 239-254, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28343375

ABSTRACT

PURPOSE: Lapatinib is a candidate drug for treatment of trastuzumab-resistant, human epidermal growth factor receptor 2 (HER2)-positive gastric cancer (GC). Unfortunately, lapatinib resistance renders this drug ineffective. The present study investigated the implication of forkhead box O1 (FOXO1) signaling in the acquired lapatinib resistance in HER2-positive GC cells. MATERIALS AND METHODS: Lapatinib-resistant GC cell lines (SNU-216 LR2-8) were generated in vitro by chronic exposure of lapatinib-sensitive, HER2-positive SNU-216 cells to lapatinib. SNU-216 LR cells with FOXO1 overexpression were generated by stable transfection of a constitutively active FOXO1 mutant (FOXO1A3). HER2 and MET in SNU-216 LR cells were downregulated using RNA interference. The sensitivity of GC cells to lapatinib and/or cisplatin was determined by crystal violet assay. In addition, Western blot analysis, luciferase reporter assay and reverse transcription-polymerase chain reaction were performed. RESULTS: SNU-216 LR cells showed upregulations of HER2 and MET, but downregulation of FOXO1 compared to parental SNU-216 cells. FOXO1 overexpression in SNU-216 LR cells significantly suppressed resistance to lapatinib and/or cisplatin. In addition, FOXO1 negatively controlled HER2 and MET at the transcriptional level and was negatively controlled by these molecules at the post-transcriptional level. A positive crosstalk was shown between HER2 and MET, each of which increased resistance to lapatinib and/or cisplatin. CONCLUSION: FOXO1 serves as an important linker between HER2 and MET signaling pathways through negative crosstalks and is a key regulator of the acquired lapatinib resistance in HER2-positive GC cells. These findings provide a rationale for establishing a novel treatment strategy to overcome lapatinib resistance in a subtype of GC patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Forkhead Box Protein O1/metabolism , Proto-Oncogene Proteins c-met/metabolism , Quinazolines/pharmacology , Receptor, ErbB-2/biosynthesis , Stomach Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Forkhead Box Protein O1/antagonists & inhibitors , Humans , Lapatinib , Proto-Oncogene Proteins c-met/genetics , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Signal Transduction , Stomach Neoplasms/enzymology , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Transfection , Up-Regulation
3.
Biochem Biophys Res Commun ; 493(3): 1349-1355, 2017 11 25.
Article in English | MEDLINE | ID: mdl-28970066

ABSTRACT

Gastric cancer (GC) is a major of cause of cancer-related death and is characterized by its heterogeneity and molecular complexity. FOXO1 is a transcription factor that plays a key role in GC growth and metastasis. However, the implication of FOXO1 in GC cell stemness has been elusive. This study, for the first time, demonstrates that FOXO1 regulates GC cell stemness in association with LGR5. FOXO1 expression was significantly lower in GC tumorsphere cells than in adherent GC cells. FOXO1 silencing and overexpression promoted and inhibited the tumorsphere formation capacity of GC cells, respectively. Additionally, there was an inverse correlation between FOXO1 and GC stem cell marker LGR5 in human GC specimens. Further in vitro and in vivo experiments showed that negative crosstalk between these two molecules exists and that LGR5 silencing reversed the FOXO1 shRNA-induced tumorsphere formation even without FOXO1 restoration. Taken together, our results suggest that FOXO1 inhibits the self-renewal capacity of GC cells through interaction with LGR5. Thus, FOXO1/LGR5 signaling pathway may provide a novel targeted therapy for GC.


Subject(s)
Forkhead Box Protein O1/metabolism , Receptors, G-Protein-Coupled/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Animals , Cell Line, Tumor , Forkhead Box Protein O1/genetics , Gene Expression Regulation, Neoplastic , Humans , Mice, Inbred BALB C , Neoplastic Stem Cells/pathology , Receptors, G-Protein-Coupled/genetics , Stomach Neoplasms/genetics , Xenograft Model Antitumor Assays
4.
Cell Death Dis ; 8(5): e2793, 2017 05 18.
Article in English | MEDLINE | ID: mdl-28518146

ABSTRACT

Acyl-CoA thioesterase 7 (ACOT7) is a major isoform of the ACOT family that catalyzes hydrolysis of fatty acyl-CoAs to free fatty acids and CoA-SH. However, canonical and non-canonical functions of ACOT7 remain to be discovered. In this study, for the first time, ACOT7 was shown to be responsive to genotoxic stresses such as ionizing radiation (IR) and the anti-cancer drug doxorubicin in time- and dose-dependent manners. ACOT7 knockdown induced cytostasis via activation of the p53-p21 signaling pathway without a DNA damage response. PKCζ was specifically involved in ACOT7 depletion-mediated cell cycle arrest as an upstream molecule of the p53-p21 signaling pathway in MCF7 human breast carcinoma and A549 human lung carcinoma cells. Of the other members of the ACOT family, including ACOT1, 4, 8, 9, 11, 12, and 13 that were expressed in human, ACOT4, 8, and 12 were responsive to genotoxic stresses. However, none of those had a role in cytostasis via activation of the PKCζ-p53-p21 signaling pathway. Analysis of the ACOT7 prognostic value revealed that low ACOT7 levels prolonged overall survival periods in breast and lung cancer patients. Furthermore, ACOT7 mRNA levels were higher in lung cancer patient tissues compared to normal tissues. We also observed a synergistic effect of ACOT7 depletion in combination with either IR or doxorubicin on cell proliferation in breast and lung cancer cells. Together, our data suggest that a low level of ACOT7 may be involved, at least in part, in the prevention of human breast and lung cancer development via regulation of cell cycle progression.


Subject(s)
Cell Cycle , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Protein Kinase C/metabolism , Signal Transduction , Thiolester Hydrolases/metabolism , Tumor Suppressor Protein p53/metabolism , A549 Cells , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Cycle Checkpoints/drug effects , DNA Damage , Down-Regulation/drug effects , Down-Regulation/radiation effects , Doxorubicin/pharmacology , Drug Synergism , Humans , MCF-7 Cells , Masoprocol/pharmacology , Radiation, Ionizing
5.
Pathobiology ; 84(4): 210-217, 2017.
Article in English | MEDLINE | ID: mdl-28399526

ABSTRACT

OBJECTIVE: Gastric cancer (GC) is the second most common cancer and the third leading cause of cancer-related death in Korea. Alterations in the ERBB (homology to the erythroblastoma viral gene product, v-erbB) receptor family and ERBB-related signaling pathways are frequently observed in GC. However, the roles of the ERBB receptors and their ligands in GC are not well established. METHODS: We evaluated the expression levels of various ERBB receptor ligands (i.e., heparin-binding epidermal growth factor-like growth factor [HBEGF], transforming growth factor-α [TGFA], amphiregulin [AREG], epiregulin [EREG], epidermal growth factor [EGF], and betacellulin [BTC]) and 3 ERBB family receptors (i.e., epidermal growth factor receptor [EGFR], human EGFR2 [HER2], and ERBB3) in 313 cases of GC using immunohistochemistry, fluorescence in situ hybridization, and mRNA in situ hybridization. RESULTS: A high expression of EGFR, HER2, and ERBB3 was observed in 30, 32, and 27 cases, respectively. A high expression of HBEGF, TGFA, AREG, EREG, EGF, and BTC was observed in 91, 97, 151, 74, 26, and 37 cases, respectively. A high expression of TGFA was associated with better survival, while a high expression of BTC was associated with worse survival. These results were confirmed using Cox proportional hazards analysis. HBEGF, TGFA, AREG, tumor-node-metastasis classification, Lauren's classification, and ERBB3 were significant survival parameters in multivariate analysis. CONCLUSION: Among the ERBB family receptors and ligands examined, 3 ligands (i.e., TGFA, HBEGF, and AREG) and ERBB3 had a prognostic impact.


Subject(s)
ErbB Receptors/metabolism , Signal Transduction , Stomach Neoplasms/metabolism , Female , Humans , In Situ Hybridization, Fluorescence , Ligands , Male , Middle Aged , Prognosis , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Republic of Korea , Stomach Neoplasms/mortality , Tissue Array Analysis
6.
World J Gastroenterol ; 22(41): 9141-9153, 2016 Nov 07.
Article in English | MEDLINE | ID: mdl-27895401

ABSTRACT

AIM: To investigated the relationships between HER2, c-Jun N-terminal kinase (JNK) and protein kinase B (AKT) with respect to metastatic potential of HER2-positive gastric cancer (GC) cells. METHODS: Immunohistochemistry was performed on tissue array slides containing 423 human GC specimens. Using HER2-positve GC cell lines SNU-216 and NCI-N87, HER2 expression was silenced by RNA interference, and the activations of JNK and AKT were suppressed by SP600125 and LY294002, respectively. Transwell assay, Western blot, semi-quantitative reverse transcription-polymerase chain reaction and immunofluorescence staining were used in cell culture experiments. RESULTS: In GC specimens, HER2, JNK, and AKT activations were positively correlated with each other. In vitro analysis revealed a positive regulatory feedback loop between HER2 and JNK in GC cell lines and the role of JNK as a downstream effector of AKT in the HER2/AKT signaling pathway. JNK inhibition suppressed migratory capacity through reversing EMT and dual inhibition of JNK and AKT induced a more profound effect on cancer cell motility. CONCLUSION: HER2, JNK and AKT in human GC specimens are positively associated with each other. JNK and AKT, downstream effectors of HER2, co-operatively contribute to the metastatic potential of HER2-positive GC cells. Thus, targeting of these two molecules in combination with HER2 downregulation may be a good approach to combat HER2-positive GC.


Subject(s)
Cell Movement , Epithelial-Mesenchymal Transition , JNK Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/metabolism , Signal Transduction , Stomach Neoplasms/enzymology , Cell Line, Tumor , Cell Movement/drug effects , Enzyme Activation , Epithelial-Mesenchymal Transition/drug effects , Feedback, Physiological , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neoplasm Metastasis , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , RNA Interference , Receptor, ErbB-2/genetics , Signal Transduction/drug effects , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Time Factors , Transfection
7.
APMIS ; 124(9): 748-56, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27365055

ABSTRACT

Since the molecular mechanism of hypoxic adaptation in cancer cells is cell-type specific, we investigated whether glycogen synthase kinase-3ß (GSK-3ß) activation is involved in hypoxia-induced gastric tumor promotion. Stable gastric cancer cell lines (SNU-638, SNU-484, MKN1, and MKN45) were cultured under hypoxic conditions. Cells overexpressing wild-type GSK-3ß (WT-GSK-3ß) or kinase-dead mutant of GSK-3ß (KD-GSK-3ß) were generated and used for cell culture and animal studies. In cell culture experiments, hypoxia decreased GSK-3ß activation in gastric cancer cells. Cell viability and the expressions of HIF-1α protein and VEGF mRNA in gastric cancer cells were higher in KD-GSK-3ß transfectants than in WT-GSK-3ß transfectants under hypoxic conditions, but not under normoxic conditions. Gastric cancer xenografts showed that tumor growth, microvessel area, HIF-1α activation, and VEGF expression were higher in KD-GSK-3ß tumors than in WT-GSK-3ß tumors in vivo. In addition, the expression of hypoxia-induced HIF-1α protein was regulated by GSK-3ß at the translational level. Our data suggest that GSK-3ß is involved in hypoxic adaptation of gastric cancer cells as an inhibitory upstream regulator of the HIF-1α/VEGF signaling pathway.


Subject(s)
Glycogen Synthase Kinase 3 beta/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia , Neovascularization, Pathologic , Signal Transduction , Stomach Neoplasms/pathology , Stomach Neoplasms/physiopathology , Animals , Blotting, Western , Cell Line, Tumor , Cell Survival , Disease Models, Animal , Heterografts , Humans , Immunohistochemistry , Male , Mice, Inbred BALB C , Mice, Nude
8.
BMC Gastroenterol ; 16(1): 59, 2016 Jun 06.
Article in English | MEDLINE | ID: mdl-27268017

ABSTRACT

BACKGROUND: Since the biological function of c-Jun N-terminal kinase (JNK) in gastric cancer remains unclear, we investigated the clinical significance of JNK activation and its association with FOXO1 activation. METHODS: Immunohistochemical tissue array analysis of 483 human gastric cancer specimens was performed, and the results of the immunostaining were quantified. The correlation between JNK activation (nuclear staining for pJNK) and clinicopathological features, the proliferation index, prognosis or FOXO1 inactivation (cytoplasmic staining for pFOXO1) was analyzed. The SNU-638 gastric cancer cell line was used for in vitro analysis. RESULTS: Nuclear staining of pJNK was found in 38 % of the gastric carcinomas and was higher in the early stages of pTNM (P < 0.001). pJNK staining negatively correlated with lymphatic invasion (P = 0.034) and positively correlated with intestinal type by Lauren's classification (P = 0.037), Ki-67-labeling index (P < 0.001), cyclin D1 (P = 0.045), cyclin E (P < 0.001) and pFOXO1 (P < 0.001). JNK activation correlated with a longer patients survival (P =0.008) and patients with a JNK-active and FOXO1-inactive tumor had a higher survival rate than the remainder of the population (P = 0.004). In vitro analysis showed that JNK inhibition by SP600125 in SNU-638 cells decreased cyclin D1 protein expression and increased FOXO1 activation. Further, JNK inhibition markedly suppressed colony formation, which was partially restored by FOXO1 shRNA expression. CONCLUSIONS: Our results indicate that JNK activation may serve as a valuable prognostic factor in gastric cancer, and that it is implicated in gastric tumorigenesis, at least in part, through FOXO1 inhibition.


Subject(s)
Forkhead Box Protein O1/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Stomach Neoplasms/metabolism , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Proliferation , Enzyme Activation , Female , Humans , Immunohistochemistry , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Male , Middle Aged , Prognosis , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Survival Rate
9.
Gastric Cancer ; 19(3): 767-77, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26386561

ABSTRACT

BACKGROUND: Cells expressing LGR5, an intestinal stem cell marker, have been suggested as cancer stem cells in human colon cancers. Previously, we discovered that LGR5-expressing cells exist in the gastric antrum and remarkably increase in number in intestinal metaplasia. In addition, most gastric adenomas contain abundant LGR5-expressing cells coexpressing intestinal stem cell signatures. However, LGR5 expression in gastric cancers (GCs) and its prognostic significance remain unknown. METHODS: We examined the LGR5 expression in GC tissues by real time-PCR and RNA in situ hybridization, and analyzed its clinicopathological relevance and prognostic value. The effects of LGR5 on cancer cell proliferation and migration were assessed with an in vitro transfection technique. RESULTS: LGR5 expression was significantly lower in GCs than in matched nontumorous gastric mucosa. RNA in situ hybridization on tissue microarrays showed that 7 % of GCs were positive for LGR5. LGR5 positivity was associated with old age, well to moderate differentiation, and nuclear ß-catenin positivity. Although LGR5 did not show any prognostic significance for all GC cases, it was associated with poor survival in GCs with nuclear ß-catenin expression. LGR5 expression was induced by transfection in GC cell lines with abnormal Wnt activation, which, however, showed no influence on the growth and migration of GC cells. CONCLUSION: A small portion of GCs expressed LGR5. Although LGR5 was associated with poor survival in GCs with nuclear ß-catenin, LGR5 expression in GC cells had no effects on the growth and migration, requiring a further study exploring a biological role of LGR5 in GCs.


Subject(s)
Adenocarcinoma/secondary , Biomarkers, Tumor/metabolism , Carcinoma, Signet Ring Cell/secondary , Neoplastic Stem Cells/metabolism , Receptors, G-Protein-Coupled/metabolism , Stomach Neoplasms/pathology , Adenocarcinoma/metabolism , Adenocarcinoma/surgery , Aged , Apoptosis , Biomarkers, Tumor/genetics , Blotting, Western , Carcinoma, Signet Ring Cell/metabolism , Carcinoma, Signet Ring Cell/surgery , Case-Control Studies , Cell Movement , Cell Proliferation , Female , Follow-Up Studies , Gastrectomy , Gastric Mucosa/metabolism , Humans , Immunoenzyme Techniques , Intestinal Mucosa/metabolism , Intestines/pathology , Lymphatic Metastasis , Male , Neoplasm Staging , Neoplastic Stem Cells/pathology , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, G-Protein-Coupled/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stomach/pathology , Stomach Neoplasms/metabolism , Stomach Neoplasms/surgery , Survival Rate , Wound Healing
10.
Cancer Res Treat ; 48(1): 345-54, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25761483

ABSTRACT

PURPOSE: We previously reported that forkhead transcription factors of the O class 1 (FOXO1) expression in gastric cancer (GC) was associated with angiogenesis-related molecules. However, there is little experimental evidence for the direct role of FOXO1 in GC. In the present study, we investigated the effect of FOXO1 on the tumorigenesis and angiogenesis in GC and its relationship with SIRT1. MATERIALS AND METHODS: Stable GC cell lines (SNU-638 and SNU-601) infected with a lentivirus containing FOXO1 shRNA were established for animal studies as well as cell culture experiments. We used xenograft tumors in nude mice to evaluate the effect of FOXO1 silencing on tumor growth and angiogenesis. In addition, we examined the association between FOXO1 and SIRT1 by immunohistochemical tissue array analysis of 471 human GC specimens and Western blot analysis of xenografted tumor tissues. RESULTS: In cell culture, FOXO1 silencing enhanced hypoxia inducible factor-1α (HIF-1α) expression and GC cell growth under hypoxic conditions, but not under normoxic conditions. The xenograft study showed that FOXO1 downregulation enhanced tumor growth, microvessel areas, HIF-1α activation and vascular endothelial growth factor (VEGF) expression. In addition, inactivated FOXO1 expression was associated with SIRT1 expression in human GC tissues and xenograft tumor tissues. CONCLUSION: Our results indicate that FOXO1 inhibits GC growth and angiogenesis under hypoxic conditions via inactivation of the HIF-1α-VEGF pathway, possibly in association with SIRT1. Thus, development of treatment modalities aiming at this pathway might be useful for treating GC.


Subject(s)
Forkhead Box Protein O1/metabolism , Neovascularization, Pathologic , Sirtuin 1/metabolism , Stomach Neoplasms/blood supply , Stomach Neoplasms/metabolism , Animals , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation , Forkhead Box Protein O1/antagonists & inhibitors , Forkhead Box Protein O1/biosynthesis , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Nude , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
11.
Br J Cancer ; 113(8): 1186-96, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26448177

ABSTRACT

BACKGROUND: The biological significance of FOXO1, a member of the forkhead box O transcription factor family, in gastric cancer (GC) remains unclear. The present study provides direct evidence of the role of FOXO1 in tumour growth and metastasis of GC in relation to human epidermal growth factor receptor 2 (HER2). METHODS: The expressions of FOXO1 and HER2 were modulated in GC cell lines (SNU-638, MKN45, SNU-216 and NCI-N87) by stable transfections. The effects of transfection on GC phenotypes were evaluated in vitro and in animal models. In addition, the relationship between FOXO1 and HER2 was analysed using GC clinical specimens, cell lines and xenografts. RESULTS: FOXO1 silencing in GC cells increased colony formation and mesenchymal transition in vitro, as well as tumour growth and metastasis in nude mice, whereas HER2 silencing induced the opposite results.. Furthermore, an inverse relationship between FOXO1 and HER2 was found in clinical specimens of GC, GC cells and GC xenograft tumours. Although a negative crosstalk between these two molecules was shown, double knockdown of both FOXO1 and HER2 in GC cells revealed that HER2 silencing reversed the FOXO1 shRNA-induced migration and invasion even without the FOXO1 restoration. CONCLUSIONS: Our results indicate that loss of FOXO1 promotes GC growth and metastasis by upregulating HER2 expression and that the HER2 expression is more critical to the induction of GC cell metastasis. The present study provides evidence that the FOXO1/HER2 pathway may regulate GC progression in a subgroup of GC patients.


Subject(s)
Cell Proliferation/genetics , Forkhead Transcription Factors/genetics , Neoplasm Metastasis/genetics , Receptor, ErbB-2/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Up-Regulation/genetics , Animals , Cell Line, Tumor , Cell Movement/genetics , Forkhead Box Protein O1 , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/genetics
12.
Virchows Arch ; 467(3): 285-94, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26070873

ABSTRACT

Olfactomedin 4 (OLFM4) has been demonstrated to be upregulated in various cancers and involved in many cellular processes such as cell adhesion, apoptosis, and cell proliferation. In gastric cancer, clinicopathological relevance of OLFM4 expression has been reported. However, there are few studies showing how expression of OLFM4 evolves during multistep gastric carcinogenesis. In this study, we investigated OLFM4 expression during gastric carcinogenesis using RNA in situ hybridization (ISH). We found that OLFM4 expression is absent in normal gastric mucosa, begins to appear at the isthmus region in gastric glands in chronic gastritis, and is remarkably increased in intestinal metaplasia (IM). Interestingly, gastric-type glands around IM frequently expressed OLFM4 before CDX2 was expressed, suggesting that OLFM4 might be involved in regulating CDX2 expression. However, overexpression of OLFM4 failed to induce CDX2 transcription. All gastric adenomas were strongly positive for OLFM4. OLFM4 expression was higher in intestinal type, well to moderately differentiated and early-stage adenocarcinomas, and decreased in poorly differentiated and advanced-stage gastric cancer (GC). Although OLFM4 expression had no prognostic value for GC overall (P = 0.441), it was associated with poor survival of GC in stage II, III, and IV (P = 0.018), suggesting that OLFM4 expression has prognostic significance for late-stage GC. Our findings suggest that OLFM4 is not only involved in early stages of gastric carcinogenesis but also a useful prognostic marker for advanced GC, which is encouraging for further studies exploring OLFM4 as a potential target for therapy of GC.


Subject(s)
Biomarkers, Tumor/analysis , Granulocyte Colony-Stimulating Factor/biosynthesis , Stomach Neoplasms/pathology , Aged , Carcinogenesis , Humans , Immunohistochemistry , In Situ Hybridization , Kaplan-Meier Estimate , Middle Aged , Prognosis , Real-Time Polymerase Chain Reaction , Stomach Neoplasms/mortality , Tissue Array Analysis
13.
PLoS One ; 10(5): e0127300, 2015.
Article in English | MEDLINE | ID: mdl-25996368

ABSTRACT

Gastric intestinal metaplasia (IM) is a highly prevalent preneoplastic lesion; however, the molecular mechanisms regulating its development remain unclear. We have previously shown that a population of cells expressing the intestinal stem cell (ISC) marker LGR5 increases remarkably in IM. In this study, we further investigated the molecular characteristics of these LGR5+ cells in IM by examining the expression profile of several ISC markers. Notably, we found that ISC markers-including OLFM4 and EPHB2-are positively associated with the CDX2 expression in non-tumorous gastric tissues. This finding was confirmed in stomach lesions with or without metaplasia, which demonstrated that OLFM4 and EPHB2 expression gradually increased with metaplastic progression. Moreover, RNA in situ hybridization revealed that LGR5+ cells coexpress several ISC markers and remained confined to the base of metaplastic glands, reminiscent to that of normal intestinal crypts, whereas those in normal antral glands expressed none of these markers. Furthermore, a large number of ISC marker-expressing cells were diffusely distributed in gastric adenomas, suggesting that these markers may facilitate gastric tumorigenesis. In addition, Barrett's esophagus (BE)-which is histologically similar to intestinal metaplasia-exhibited a similar distribution of ISC markers, indicating the presence of a stem cell population with intestinal differentiation potential. In conclusion, we identified that LGR5+ cells in gastric IM and BE coexpress ISC markers, and exhibit the same expression profile as those found in normal intestinal crypts. Taken together, these results implicate an intestinal-like stem cell population in the pathogenesis of IM, and provide an important basis for understanding the development and maintenance of this disease.


Subject(s)
Barrett Esophagus/metabolism , Barrett Esophagus/pathology , Gastric Mucosa/metabolism , Intestinal Mucosa/metabolism , Stem Cells/metabolism , Stomach/pathology , Adenoma/genetics , Adenoma/metabolism , Adenoma/pathology , Barrett Esophagus/genetics , Biomarkers , CDX2 Transcription Factor , Disease Progression , Gene Expression , Homeodomain Proteins/metabolism , Humans , Intestines/pathology , Metaplasia , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
14.
APMIS ; 123(5): 373-82, 2015 May.
Article in English | MEDLINE | ID: mdl-25846563

ABSTRACT

The transcription factor signal transducers and activators of transcription 3 (STAT3) can promote cancer metastasis, but its underlying regulatory mechanisms in gastric cancer cell invasiveness still remain obscure. We investigated the relationship between STAT3 and glycogen synthase kinase-3ß (GSK-3ß) and its significance in metastatic potential in gastric cancer cells. Immunohistochemical tissue array analysis of 267 human gastric carcinoma specimens showed that the expressions of active forms of STAT3 (pSTAT3) and GSK-3ß (pGSK-3ß) were found in 68 (25%) and 124 (46%) of 267 gastric cancer cases, respectively, showing a positive correlation (p < 0.001). Cell culture experiments using gastric cancer cell lines SNU-638 and SNU-668 revealed that STAT3 suppression did not affect pGSK-3ß expression, whereas GSK-3ß inhibition reduced pSTAT3 expression. With respect to metastatic potential in gastric cancer cells, both STAT3 suppression and GSK-3ß inhibition decreased cell migration, invasion, and mesenchymal marker (Snail, Vimentin, and MMP9) expression. Moreover, the inhibitory effects of STAT3 and GSK-3ß on cell migration were synergistic. These results demonstrated that STAT3 and GSK-3ß are positively associated and synergistically contribute to metastatic potential in gastric cancer cells. Thus, dual use of STAT3 and GSK-3ß inhibitors may enhance the efficacy of the anti-metastatic treatment of gastric cancer.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , STAT3 Transcription Factor/metabolism , Stomach Neoplasms/metabolism , Amino Acid Substitution , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Movement/physiology , Epithelial-Mesenchymal Transition/physiology , Gene Knockdown Techniques , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Humans , Immunohistochemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Neoplasm Invasiveness/physiopathology , RNA, Small Interfering/genetics , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , Signal Transduction , Stomach Neoplasms/pathology , Stomach Neoplasms/secondary , Thiazoles/pharmacology , Tyrphostins/pharmacology , Urea/analogs & derivatives , Urea/pharmacology
15.
J Cancer Res Clin Oncol ; 141(7): 1181-8, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25475007

ABSTRACT

PURPOSE: Bile acids might induce mucin expression and regulate tumor behavior in esophageal and colon cancers. However, little is known about the effect of bile acids on tumor invasiveness of gastric carcinoma (GC). The aim of the current study was to elucidate the mechanisms by which bile acids regulate tumor invasion in GC. METHODS: We investigated bile acid-induced MUC2 expression and cell invasion and migration in the cultured GC cell lines, SNU-216, and MKN45. In addition, immunohistochemical analysis of MUC2 and Snail was performed on 389 archival paraffin-embedded tissues of GC to evaluate the correlation of their expression with prognosis. RESULTS: Deoxycholic acid (DCA), a secondary bile acid, had no effect on the viability of SNU-216 and MKN45 GC cells at low concentrations (0-100 µM), but decreased viability at a higher concentration (200 µM). MKN45 cells showed higher MUC2 expression than SNU-216 cells under basal conditions. DCA treatment upregulated MUC2 mRNA expression in both SNU-216 and MKN45 cells. Expression of Snail and MMP9 was markedly decreased by DCA treatment, and E-cadherin expression was subsequently increased. DCA significantly inhibited invasion and migration of SNU-216 and MKN45 cells. In human GC, MUC2 expression showed a negative correlation with Snail expression (P = 0.021) and a significantly positive correlation with better prognosis (P = 0.023). CONCLUSIONS: Taken together, our data suggest that DCA induced MUC2 expression in GC cells and inhibited tumor invasion and migration. Additionally, MUC2-expressing GCs showed low rates of Snail expression and were associated with a favorable prognosis.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/pathology , Deoxycholic Acid/pharmacology , Mucin-2/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Adenocarcinoma/diagnosis , Bile Acids and Salts/pharmacology , Biomarkers, Tumor/genetics , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mucin-2/metabolism , Neoplasm Invasiveness , Prognosis , Stomach Neoplasms/diagnosis , Tissue Array Analysis , Tumor Cells, Cultured
16.
APMIS ; 122(9): 848-55, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24673584

ABSTRACT

FOXO1, a forkhead box O (FOXO) transcription factor, and nuclear factor-κB (NF-κB) are prognostically significant transcription factors in gastric cancer. As their relationship has been inconsistent depending on the cell type, we aimed to investigate whether FOXO1 is associated with NF-κB p65 (RelA) in gastric cancer. Immunohistochemistry was performed on tissue array slides containing 298 gastric carcinoma specimens. We found that the cytoplasmic expression of pFOXO1, the inactive form of FOXO1, was positively correlated with nuclear RelA expression (p = 0.024). In addition, the expressions of pFOXO1 and RelA were positively related with cyclin D1 expression (p = 0.014 and p = 0.001, respectively) and Ki-67 labeling index (p = 0.025 and p = 0.017, respectively). However, they did not show association with the expressions of cyclin E, p53 and pRb. Cell culture experiments showed that FOXO1 overexpression by transfection of FOXO1 AAA mutant gene suppressed NF-κB activation in SNU-484 gastric cancer cells. These results suggest that FOXO1 and NF-κB are negatively associated and that FOXO1 is a negative upstream regulator of NF-κB in gastric cancer.


Subject(s)
Forkhead Transcription Factors/metabolism , Stomach Neoplasms/pathology , Transcription Factor RelA/antagonists & inhibitors , Cell Line, Tumor , Cyclin D1/biosynthesis , Cyclin E/biosynthesis , Forkhead Box Protein O1 , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Ki-67 Antigen , Mutation , Retinoblastoma Protein/biosynthesis , Transcription Factor RelA/biosynthesis , Tumor Suppressor Protein p53/biosynthesis
17.
Pathobiology ; 81(1): 25-35, 2014.
Article in English | MEDLINE | ID: mdl-23969480

ABSTRACT

OBJECTIVES: The aims of this study were to assess expressions of the DNA damage response (DDR)-related proteins and to investigate their clinical significances in gastric carcinoma. METHODS: Two independent cohorts, a training set (n=524) and validation set (n=394), of gastric cancer patients were enrolled. Ataxia telangiectasia mutated (ATM), checkpoint kinase 2 (Chk2), and p53 expressions were examined by immunohistochemistry using tissue microarray. RESULTS: ATM loss, Chk2 loss, and p53 positivity were observed in 21.8, 14.1, and 36.1% of the training set, and in 17.3, 12.2, and 35.8% of the validation set, respectively. In the training set, the aberrant expressions of ATM, Chk2, or p53 were significantly associated with an advanced TNM stage and poor disease-specific survival. This association was verified in the validation set. Chk2 positivity and p53 negativity were significantly related to a prolonged disease-specific survival. Also, patients with nonaberrant expressional levels of all 3 DDR-related proteins had a more favorable outcome than others. Multivariate analyses showed that Chk2 loss and at least 1 aberrant DDR-related protein remained as independent prognostic factors of poor disease-specific survival. CONCLUSIONS: This study elucidated the prognostic implications of DDR-related proteins, and suggests that their aberrant expressions play critical roles in the development and progression of gastric cancer.


Subject(s)
Adenocarcinoma/metabolism , Ataxia Telangiectasia Mutated Proteins/biosynthesis , Checkpoint Kinase 2/biosynthesis , DNA Damage , Stomach Neoplasms/metabolism , Tumor Suppressor Protein p53/biosynthesis , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Adult , Aged , Ataxia Telangiectasia Mutated Proteins/analysis , Biomarkers, Tumor/analysis , Checkpoint Kinase 2/analysis , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Staging , Prognosis , Proportional Hazards Models , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Tissue Array Analysis , Tumor Suppressor Protein p53/analysis
18.
Gastric Cancer ; 17(3): 423-30, 2014.
Article in English | MEDLINE | ID: mdl-24202965

ABSTRACT

BACKGROUND: Cisplatin (CDDP) is one of the most important chemotherapeutic agents in the treatment of advanced gastric cancer, but its efficacy is limited by CDDP resistance. Because the transcription factor FOXO1 is related to chemoresistance in various cancer cells, we investigated the function of FOXO1 in CDDP resistance in human gastric cancer cells. METHODS: Human gastric cancer cell lines MKN45 and SNU-601 were used. FOXO1 activation was modulated by transfection of FOXO1 AAA mutant gene or FOXO1 shRNA. The effects of FOXO1 on cell growth and CDDP cytotoxicity were assessed by crystal violet assay. Protein expressions of FOXO1, p110α, pAkt, and Akt were analyzed by Western blotting, and FOXO1 mRNA expression was evaluated by semiquantitative reverse transcription-polymerase chain reaction. FOXO1 activity was determined by luciferase reporter assay, and cell apoptosis was assessed by DAPI staining and Western blotting for PARP cleavage. RESULTS: Cisplatin treatment induced FOXO1 expression and activation in both gastric cancer cell lines. FOXO1 overexpression increased the CDDP resistance without changes in cell growth, whereas FOXO1 silencing enhanced CDDP cytotoxicity along with apoptotic characteristics. Both constitutive and CDDP-induced FOXO1 activations were accompanied by an increase in p110α and pAkt expression. Furthermore, Akt inhibition by LY294002 treatment restored the CDDP cytotoxicity that was suppressed by FOXO1 overexpression. CONCLUSION: FOXO1 inhibits CDDP-induced apoptosis in gastric cancer cells via activating PI3K/Akt pathway. Thus, FOXO1 may be an useful pharmacological indicator to predict CDDP efficacy in gastric cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Forkhead Transcription Factors/genetics , Stomach Neoplasms/drug therapy , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Forkhead Box Protein O1 , Humans , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
19.
PLoS One ; 8(12): e82390, 2013.
Article in English | MEDLINE | ID: mdl-24340024

ABSTRACT

Lgr5 was identified as a promising gastrointestinal tract stem cell marker in mice. Lineage tracing indicates that Lgr5(+) cells may not only be the cells responsible for the origin of tumors; they may also be the so-called cancer stem cells. In the present study, we investigated the presence of Lgr5(+) cells and their biological significance in normal human gastric mucosa and gastric tumors. RNAscope, a newly developed RNA in situ hybridization technique, specifically labeled Lgr5(+) cells at the basal glands of the gastric antrum. Notably, the number of Lgr5(+) cells was remarkably increased in intestinal metaplasia. In total, 76% of gastric adenomas and 43% of early gastric carcinomas were positive for LGR5. Lgr5(+) cells were found more frequently in low-grade tumors with active Wnt signaling and an intestinal gland type, suggesting that LGR5 is likely involved in the very early stages of Wnt-driven tumorigenesis in the stomach. Interestingly, similar to stem cells in normal tissues, Lgr5(+) cells were often restricted to the base of the tumor glands, and such Lgr5(+) restriction was associated with high levels of intestinal stem cell markers such as EPHB2, OLFM4, and ASCL2. Thus, our findings show that Lgr5(+) cells are present at the base of the antral glands in the human stomach and that this cell population significantly expands in intestinal metaplasias. Furthermore, Lgr5(+) cells are seen in a large number of gastric tumors ; their frequent basal arrangements and coexpression of ISC markers support the idea that Lgr5(+) cells act as stem cells during the early stage of intestinal-type gastric tumorigenesis.


Subject(s)
Neoplasm Proteins/metabolism , Neoplastic Stem Cells , Receptors, G-Protein-Coupled/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Animals , Female , Humans , Male , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Receptors, G-Protein-Coupled/genetics , Stomach Neoplasms/genetics
20.
J Virol ; 87(19): 10515-23, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23824821

ABSTRACT

In Epstein-Barr virus (EBV)-infected gastric carcinoma, EBV-encoded BARF1 has been hypothesized to function as an oncogene. To evaluate cellular changes induced by BARF1, we isolated the full-length BARF1 gene from gastric carcinoma cells that were naturally infected with EBV and transfected BARF1 into EBV-negative gastric carcinoma cells. BARF1 protein was primarily secreted into culture supernatant and only marginally detectable within cells. Compared with gastric carcinoma cells containing empty vector, BARF1-expressing gastric carcinoma cells exhibited increased cell proliferation (P < 0.05). There were no significant differences in apoptosis, invasion, or migration between BARF1-expressing gastric carcinoma cells and empty vector-transfected cells. BARF1-expressing gastric carcinoma cells demonstrated increased nuclear expression of nuclear factor kappa B (NF-κB) RelA protein and increased NF-κB-dependent cyclin D1. The expression of p21(WAF1) was diminished by BARF1 transfection and increased by NF-κB inhibition. Proliferation of naturally EBV-infected gastric carcinoma cells was suppressed by BARF1 small interfering RNA (siRNA) (P < 0.05). Immunohistochemical analysis of 120 human gastric carcinoma tissues demonstrated increased expression of cyclin D1 and reduced expression of p21(WAF1) in EBV-positive samples versus EBV-negative gastric carcinomas (P < 0.05). In conclusion, the secreted BARF1 may stimulate proliferation of EBV-infected gastric carcinoma cells via upregulation of NF-κB/cyclin D1 and reduction of the cell cycle inhibitor p21(WAF1), thereby facilitating EBV-induced cancer progression.


Subject(s)
Cell Proliferation , Epstein-Barr Virus Infections/pathology , Herpesvirus 4, Human/pathogenicity , NF-kappa B/metabolism , Stomach Neoplasms/pathology , Viral Proteins/metabolism , Apoptosis , Blotting, Western , Cell Adhesion , Cell Movement , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/virology , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , NF-kappa B/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/metabolism , Stomach Neoplasms/virology , Tumor Cells, Cultured , Viral Proteins/antagonists & inhibitors , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...