Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
J Exp Clin Cancer Res ; 38(1): 127, 2019 Mar 12.
Article in English | MEDLINE | ID: mdl-30866995

ABSTRACT

BACKGROUND: Mechanistic target of rapamycin (mTOR) pathway is essential for the growth of gastric cancer (GC), but mTOR inhibitor everolimus was not effective for the treatment of GCs. The Cancer Genome Atlas (TCGA) researchers reported that most diffuse-type GCs were genomically stable (GS). Pathological analysis suggested that some diffuse-type GCs developed from intestinal-type GCs. METHODS: We established patient-derived xenograft (PDX) lines from diffuse-type GCs, and searched for drugs that suppressed their growth. Diffuse-type GCs were classified into subtypes by their gene expression profiles. RESULTS: mTOR inhibitor temsirolimus strongly suppressed the growth of PDX-derived diffuse-type GC-initiating cells, which was regulated via Wnt-mTOR axis. These cells were microsatellite unstable (MSI) or chromosomally unstable (CIN), inconsistent with TCGA report. Diffuse-type GCs in TCGA cohort could be classified into two clusters, and GS subtype was major in cluster I while CIN and MSI subtypes were predominant in cluster II where PDX-derived diffuse-type GC cells were included. We estimated that about 9 and 55% of the diffuse-type GCs in cluster II were responders to mTOR inhibitors and checkpoint inhibitors, respectively, by identifying PIK3CA mutations and MSI condition in TCGA cohort. These ratios were far greater than those of diffuse-type GCs in cluster I or intestinal-type GCs. Further analysis suggested that diffuse-type GCs in cluster II developed from intestinal-type GCs while those in cluster I from normal gastric epithelial cells. CONCLUSION: mTOR inhibitors and checkpoint inhibitors might be useful for the treatment of a subset of diffuse-type GCs which may develop from intestinal-type GCs.


Subject(s)
Stomach Neoplasms/drug therapy , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Humans , Mice , Microsatellite Instability , Stomach Neoplasms/pathology
2.
Br J Cancer ; 118(7): 972-984, 2018 04.
Article in English | MEDLINE | ID: mdl-29527007

ABSTRACT

BACKGROUND: Diffuse-type gastric cancer (DGC) exhibits rapid disease progression and poor patient prognosis. We have previously established an E-cadherin/p53 double conditional knockout (DCKO) mouse line as the first genetically engineered one, which morphologically and molecularly recapitulates human DGC. In this study, we explored low-molecular-weight drugs selectively eliminating mouse and human DGC cells. METHODS: We derived mouse gastric cancer (GC) cell lines from DGC of the DCKO mice demonstrating enhanced tumourigenic activity in immunodeficient mice and acquired tolerance to cytotoxic anti-cancer agents. RESULTS: We performed a synthetic lethal screening of 1535 annotated chemical compounds, and identified 27 candidates selectively killing the GC cell lines. The most potent drug mestranol, an oestrogen derivative, and other oestrogen receptor modulators specifically attenuated cell viability of the GC cell lines by inducing apoptosis preceded by DNA damage. Moreover, mestranol could significantly suppress tumour growth of the GC cells subcutaneously transplanted into nude mice, consistent with longer survival time in the female DCKO mice than in the male. Expectedly, human E-cadherin-mutant and -low gastric cancer cells showed higher susceptibility to oestrogen drugs in contrast to E-cadherin-intact ones in vitro and in vivo. CONCLUSIONS: These findings may lead to the development of novel therapeutic strategies targeting DGC.


Subject(s)
Antineoplastic Agents/pharmacology , Stomach Neoplasms/drug therapy , Animals , Antineoplastic Agents/classification , Antineoplastic Agents/therapeutic use , Cdh1 Proteins/genetics , Cell Line, Tumor , Disease Models, Animal , Drug Screening Assays, Antitumor , Male , Mice , Mice, Knockout , Mice, Nude , Stomach Neoplasms/pathology , Tumor Suppressor Protein p53/genetics
3.
J Cancer Res Clin Oncol ; 143(3): 551-554, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27909811

ABSTRACT

PURPOSE: Diffuse-type gastric cancer (DGC) carries a poor prognosis. Effective screening is one measure that might improve the prognosis of this disease. An E-cadherin/p53 double-conditional knockout (DCKO) mouse line recapitulates human DGC morphologically and molecularly. Three circulating microRNAs (miRNA) (miR-103, miR-107, miR-194) in DCKO mice have been identified as biomarkers for DGC. We sought to evaluate whether these circulating miRNAs could be used for the detection of human DGC. METHODS: Subjects were 50 patients with DGC. Controls were first-time outpatients at Aichi Cancer Center Hospital, age- and sex-matched, without a cancer diagnosis. Total RNA containing miRNA was extracted from the plasma samples and then reverse-transcribed. The levels of miRNAs in plasma samples were quantitatively determined by real-time RT-PCR. Spiked-in cel-miR-39 was analyzed as a normalization control. RESULTS: Levels of the three plasma microRNA levels in DGC cases with or without an intestinal component were not significantly different from those in control subjects. The areas under the receiver operating characteristic curve of miR-103, miR-107, and miR-194 were 0.548, 0.563, and 0.512, respectively. CONCLUSIONS: In contrast to the DCKO mouse model, plasma miR-103, miR-107, and miR-194 levels are not altered in DGC and are not suitable for human DGC screening.


Subject(s)
Biomarkers, Tumor/blood , MicroRNAs/blood , Stomach Neoplasms/blood , Adult , Aged , Animals , Biomarkers, Tumor/genetics , Cadherins/genetics , Disease Models, Animal , Early Detection of Cancer , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Mice , Mice, Knockout , MicroRNAs/genetics , Middle Aged , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Tumor Suppressor Protein p53/genetics
4.
Oncotarget ; 7(41): 67251-67265, 2016 Oct 11.
Article in English | MEDLINE | ID: mdl-27572307

ABSTRACT

SETDB2 is a histone H3 lysine 9 (H3K9) tri-methyltransferase that is involved in transcriptional gene silencing. Since it is still unknown whether SETDB2 is linked to carcinogenesis, we studied alterations and functions of SETDB2 in human gastric cancers (GCs). SETDB2 protein was highly expressed in 30 of 72 (41.7%) primary GC tissues compared with their normal counterparts by immunohistochemistry. SETDB2 overexpression was significantly associated with the late stage of GCs (P<0.05) and poor prognosis of GC patients (P<0.05). The GC cell lines with SETDB2 knockdown and overexpression significantly decreased and increased cell proliferation, migration and invasion, respectively (P<0.05). Knockdown of SETDB2 in MKN74 and MKN45 cells reduced global H3K9 tri-methylation (me3) levels. Microarray analysis indicated that expression of WWOX and CADM1, tumor suppressor genes, was significantly enhanced in MKN74 cells after SETDB2 knockdown. Chromatin immunoprecipitation assays showed that the H3K9me3 levels at the promoter regions of these two genes corresponded to the SETDB2 expression levels in GC cells. Moreover, ectopic SETDB2 protein was recruited to their promoter regions. Our data suggest that SETDB2 is associated with transcriptional repression of WWOX and CADM1, and hence overexpression of SETDB2 may contribute to GC progression.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology , Adult , Aged , Cell Movement/physiology , Cell Proliferation/physiology , DNA Methylation , Female , Gene Expression Regulation, Neoplastic/physiology , Histones , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Stomach Neoplasms/mortality
5.
Oncotarget ; 7(4): 3966-83, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26701885

ABSTRACT

SET7/9, a histone methyltransferase, has two distinct functions for lysine methylation. SET7/9 methylates non-histone proteins, such as p53, and participates in their posttranslational modifications. Although SET7/9 transcriptionally activate the genes via H3K4 mono-methylation, its target genes are poorly understood. To clarify whether or not SET7/9 is related to carcinogenesis, we studied alterations of SET7/9 in gastric cancers (GCs). Among the 376 primary GCs, 129 cases (34.3%) showed loss or weak expression of SET7/9 protein compared to matched non-cancerous tissues by immunohistochemistry. Reduced SET7/9 expression was significantly correlated with clinical aggressiveness and worse prognosis. Knockdown of SET7/9 in GC cells markedly increased cell proliferation, migration and invasion. Expression of SREK1IP1, PGC and CCDC28B were inhibited in GC cells with SET7/9 knockdown, while matrix metalloproteinase genes (MMP1, MMP7 and MMP9) were activated. SET7/9 bound and mono-methylated H3K4 at the region of the approximately 4-6 kb upstream from the SREK1IP1 transcriptional start site and the promoters of PGC and CDC28B. Cell proliferation, migration and invasion, and expression of three MMPs were increased in GC cells with SREK1IP knockdown, which were similar to those of SET7/9 knockdown. These data suggest that SET7/9 has tumor suppressor functions, and loss of SET7/9 may contribute to gastric cancer progression.


Subject(s)
DNA Methylation , Histone-Lysine N-Methyltransferase/metabolism , Mutation/genetics , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology , Apoptosis , Blotting, Western , Cell Proliferation , Chromatin Immunoprecipitation , Disease Progression , Female , Histone-Lysine N-Methyltransferase/genetics , Humans , Immunoenzyme Techniques , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/genetics , Tumor Cells, Cultured
6.
PLoS One ; 10(12): e0145630, 2015.
Article in English | MEDLINE | ID: mdl-26695186

ABSTRACT

Twist1 overexpression is frequently observed in various cancers including gastric cancer (GC). Although DNA methylation of the Twist1 gene has been reported in cancer cells, the mechanisms underlying transcriptional activation remain uncertain. In this study, we first examined epigenetic alterations of the Twist1 using Twist1 transcription-positive and -negative cell lines that are derived from our established diffuse-type GC mouse model. Treatment with a DNA demethylation agent 5-aza-dC re-activated Twist1 expression in Twist1 expression-negative GC cells. According to methylation-specific PCR and bisulfite sequencing analysis, methylation at the CpG-rich region within Twist1 coding exon 1, rather than its promoter region, was tightly linked to transcriptional silencing of the Twist1 expression in mouse GC cells. Chromatin immunoprecipitation assays revealed that active histone mark H3K4me3 was enriched in Twist1 expression-positive cells, and inactive histone mark H3K9me3 was enriched in Twist1 expression-negative cells. The expression levels of Suv39h1 and Suv39h2, histone methyltransferases for H3K9me3, were inversely correlated with Twist1 expression, and knockdown of Suv39h1 or Suv39h2 induced Twist1 expression. Moreover, Sp1 transcription factor bound to the exon 1 CpG-rich region in Twist1 expression-positive cell lines, and Twist1 expression was diminished by mithramycin, which that interferes with Sp1 binding to CpG-rich regulatory sequences. Our studies suggested that the Twist1 transcription in GC cells might be regulated through potential cooperation of DNA methylation, histone modification in complex with Sp1 binding to CpG-rich regions within the exon 1 region.


Subject(s)
DNA Methylation , DNA, Neoplasm/metabolism , Exons , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/biosynthesis , Neoplasms, Experimental/metabolism , Nuclear Proteins/biosynthesis , Stomach Neoplasms/metabolism , Twist-Related Protein 1/biosynthesis , Animals , CpG Islands , DNA, Neoplasm/genetics , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Nuclear Proteins/genetics , Plicamycin/pharmacology , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Twist-Related Protein 1/genetics
7.
Cancer Epidemiol Biomarkers Prev ; 23(10): 2019-26, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25086101

ABSTRACT

BACKGROUND: To evaluate the relationship between methylation status of blood leukocyte DNA and risk of gastric cancer, a population-based study was conducted in Linqu County. METHODS: Methylation levels of IGFII and N33 were determined by quantitative methylation-specific PCR. The temporal trend of methylation levels during gastric cancer development was investigated in 133 gastric cancer cases from two cohorts with pre- and/or post-gastric cancer samples. As the references of pre-GCs, 204 intestinal metaplasia (IM) or dysplasia (DYS) subjects who did not progress to gastric cancer during the follow-up period were selected. Meanwhile, 285 subjects with superficial gastritis/chronic atrophic gastritis (SG/CAG) were also selected as controls. RESULTS: IGFII median methylation level was significantly higher in gastric cancer cases than those with SG/CAG (61.47% vs. 49.73%; P < 0.001). IGFII and N33 methylation levels were elevated at least 5 years ahead of clinical gastric cancer diagnosis comparing with SG/CAG (63.38% vs. 49.73% for IGFII, 9.12% vs. 5.70% for N33; all P < 0.001). Furthermore, the frequency of hypermethylated IGFII was markedly increased in IM or DYS subjects who progressed to gastric cancer in contrast to those who remained with IM and DYS, and adjusted ORs were 12.52 [95% confidence interval (CI), 3.81-41.15] for IM and 10.12 (95% CI, 2.68-38.22) for DYS. Similar results were also found for N33 in subjects with IM (OR, 3.77; 95% CI, 1.20-11.86). CONCLUSIONS: Our findings suggested that hypermethylated IGFII and N33 in blood leukocyte DNA were associated with risk of gastric cancer in a Chinese population. IMPACT: IGFII and N33 methylation status may be related to gastric carcinogenesis.


Subject(s)
Asian People/genetics , DNA Methylation/genetics , Insulin-Like Growth Factor II/genetics , Membrane Proteins/genetics , Precancerous Conditions/genetics , Stomach Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Aged , Biomarkers, Tumor/genetics , Cohort Studies , Disease Progression , Female , Humans , Leukocytes/metabolism , Male , Middle Aged , Polymerase Chain Reaction , Stomach Neoplasms/blood
8.
Clin Cancer Res ; 20(17): 4598-612, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25009298

ABSTRACT

PURPOSE: Metastasis is the leading cause of death for gastric carcinoma. An epigenetic biomarker panel for predicting gastric carcinoma metastasis could have significant clinical impact on the care of patients with gastric carcinoma. The main purpose of this study is to characterize the methylation differences between gastric carcinomas with and without metastasis. EXPERIMENTAL DESIGN: Genome-wide DNA methylation profiles between 4 metastatic and 4 nonmetastatic gastric carcinomas and their surgical margins (SM) were analyzed using methylated-CpG island amplification with microarray. The methylation states of 73 candidate genes were further analyzed in patients with gastric carcinoma in a discovery cohort (n=108) using denatured high performance liquid chromatography, bisulfite-sequencing, and MethyLight. The predictive values of potential metastasis-methylation biomarkers were validated in cohorts of patients with gastric carcinoma in China (n=330), Japan (n=129), and Korea (n=153). RESULTS: The gastric carcinoma genome showed significantly higher proportions of hypomethylation in the promoter and exon-1 regions, as well as increased hypermethylation of intragenic fragments when compared with SMs. Significant differential methylation was validated in the CpG islands of 15 genes (P<0.05) and confirmed using bisulfite sequencing. These genes included BMP3, BNIP3, CDKN2A, ECEL1, ELK1, GFRA1, HOXD10, KCNH1, PSMD10, PTPRT, SIGIRR, SRF, TBX5, TFPI2, and ZNF382. Methylation changes of GFRA1, SRF, and ZNF382 resulted in up- or downregulation of their transcription. Most importantly, the prevalence of GFRA1, SRF, and ZNF382 methylation alterations was consistently and coordinately associated with gastric carcinoma metastasis and the patients' overall survival throughout discovery and validation cohorts in China, Japan, and Korea. CONCLUSION: Methylation changes of GFRA1, SRF, and ZNF382 may be a potential biomarker set for prediction of gastric carcinoma metastasis.


Subject(s)
Carcinoma/genetics , DNA-Binding Proteins/genetics , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Serum Response Factor/genetics , Stomach Neoplasms/genetics , Transcription Factors/genetics , Adult , Aged , Biomarkers, Tumor , China , DNA Methylation/genetics , Female , Gene Expression Regulation, Neoplastic , Genome, Human , Humans , Male , Middle Aged , Neoplasm Metastasis , Promoter Regions, Genetic , Stomach Neoplasms/pathology
9.
PLoS One ; 9(2): e88347, 2014.
Article in English | MEDLINE | ID: mdl-24533079

ABSTRACT

Ovarian cancer is one of the leading causes of female death and the development of novel therapeutic approaches is urgently required. Nuclear factor-κB (NF-κB) is constitutively activated in several types of cancer including ovarian cancer and is known to support the survival of cancer cells. However, molecular mechanisms of persistent activation of NF-κB in ovarian cancer remain largely unknown. We report here that, in addition to the previously reported canonical activation, NF-κB is activated through the noncanonical pathway in ovarian cancer cells. RNA interference-mediated silencing of NF-κB inducing kinase (NIK), a central regulator of the noncanonical pathway, reduced the NF-κB2/p52 DNA binding activity and NF-κB-dependent reporter gene expression as well as NF-κB target gene expression. Notably, anchorage-dependent and -independent cell growth was impaired in NIK-depleted cells. Depletion of NIK also suppressed tumor formation in the nude mouse xenograft assay. These results indicate that NIK plays a key role in constitutive NF-κB activation and the progression of ovarian cancer cells and suggest that NIK represents an attractive therapeutic target for ovarian cancer.


Subject(s)
Disease Progression , Ovarian Neoplasms/metabolism , Protein Serine-Threonine Kinases/physiology , Animals , Apoptosis , Cell Adhesion , Cell Line, Tumor , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Silencing , Genes, Reporter , HEK293 Cells , Humans , Mice , NF-kappa B p52 Subunit/physiology , Neoplasm Transplantation , RNA Interference , Signal Transduction , NF-kappaB-Inducing Kinase
10.
Dev Growth Differ ; 55(9): 786-91, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24299059

ABSTRACT

The importance of epithelial-mesenchymal interaction on the development of gastro-intestinal (GI) organs has been repeatedly reported, but its molecular mechanism has not been fully understood though several factors including hepatocyte growth factor and endothelin-3 have been shown to mediate it. Activins have been demonstrated to play important roles in the regulation of organogenesis in vertebrates, but their roles in the regulation of growth and differentiation of GI organs remain to be solved. In the present study, we examined expression of activins in developing rat GI tract, and found that inhibin bA encoding activin A was specifically expressed by GI mesenchymes, while inhibin bB encoding activin B was expressed by both epithelial and mesenchymal components. We then examined the effect of activin A on the growth of fetal rat GI epithelial cells in primary culture. We found that activin A inhibited the growth of forestomach and glandular stomach epithelial cells while it stimulated the growth of colonic epithelial cells. These results suggest that activin A secreted from GI mesenchymes region-specifically regulates the growth of attaching epithelial cells. We thus conclude that activin A mediates epithelial-mesenchymal interaction in the developing GI tract.


Subject(s)
Cell Proliferation/physiology , Epithelial Cells/physiology , Epithelial-Mesenchymal Transition/physiology , Gastrointestinal Tract/cytology , Inhibin-beta Subunits/metabolism , Animals , Cells, Cultured , DNA Primers/genetics , Epithelial Cells/metabolism , Gastrointestinal Tract/growth & development , Rats , Reverse Transcriptase Polymerase Chain Reaction
11.
PLoS One ; 8(8): e72438, 2013.
Article in English | MEDLINE | ID: mdl-24015244

ABSTRACT

Identification of gastric tumor-initiating cells (TICs) is essential to explore new therapies for gastric cancer patients. There are reports that gastric TICs can be identified using the cell surface marker CD44 and that they form floating spheres in culture, but we could not obtain consistent results with our patient-derived tumor xenograft (PDTX) cells. We thus searched for another marker for gastric TICs, and found that CD49f(high) cells from newly-dissected gastric cancers formed tumors with histological features of parental ones while CD49f(low) cells did not when subcutaneously injected into immunodeficient mice. These results indicate that CD49f, a subunit of laminin receptors, is a promising marker for human gastric TICs. We established a primary culture system for PDTX cells where only CD49f(high) cells could grow on extracellular matrix (ECM) to form ECM-attaching spheres. When injected into immunodeficient mice, these CD49f(high) sphere cells formed tumors with histological features of parental ones, indicating that only TICs could grow in the culture system. Using this system, we found that some sphere-forming TICs were more resistant than gastric tumor cell lines to chemotherapeutic agents, including doxorubicin, 5-fluorouracil and doxifluridine. There was a patient-dependent difference in the tumorigenicity of sphere-forming TICs and their response to anti-tumor drugs. These results suggest that ECM plays an essential role for the growth of TICs, and that this culture system will be useful to find new drugs targeting gastric TICs.


Subject(s)
Biomarkers, Tumor/metabolism , Integrin alpha6/metabolism , Neoplastic Stem Cells/metabolism , Spheroids, Cellular/metabolism , Stomach Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Floxuridine/pharmacology , Fluorouracil/pharmacology , Humans , Inhibitory Concentration 50 , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Neoplasm Transplantation , Stomach Neoplasms/pathology , Tumor Cells, Cultured
12.
PLoS One ; 8(5): e62589, 2013.
Article in English | MEDLINE | ID: mdl-23667495

ABSTRACT

MicroRNAs (miRNAs) act as transcriptional regulators and play pivotal roles in carcinogenesis. According to miRNA target databases, one miRNA may regulate many genes as its targets, while one gene may be targeted by many miRNAs. These findings indicate that relationships between miRNAs and their targets may not be one-to-one. However, many reports have described only a one-to-one, one-to-multiple or multiple-to-one relationship between miRNA and its target gene in human cancers. Thus, it is necessary to determine whether or not a combination of some miRNAs would regulate multiple targets and be involved in carcinogenesis. To find some groups of miRNAs that may synergistically regulate their targets in human gastric cancer (GC), we re-analyzed our previous miRNA expression array data and found that 50 miRNAs were up-regulated on treatment with 5-aza-2'-deoxycytidine in a GC cell line. The "TargetScan" miRNA target database predicted that some of these miRNAs have common target genes. We also referred to the GEO database for expression of these common target genes in human GCs, which might be related to gastric carcinogenesis. In this study, we analyzed two miRNA combinations, miR-224 and -452, and miR-181c and -340. Over-expression of both miRNA combinations dramatically down-regulated their target genes, DPYSL2 and KRAS, and KRAS and MECP2, respectively. These miRNA combinations synergistically decreased cell proliferation upon transfection. Furthermore, we revealed that these miRNAs were down-regulated through promoter hypermethylation in GC cells. Thus, it is likely that the relationships between miRNAs and their targets are not one-to-one but multiple-to-multiple in GCs, and that these complex relationships may be related to gastric carcinogenesis.


Subject(s)
Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/genetics , Proto-Oncogene Proteins/metabolism , Stomach Neoplasms/genetics , Azacitidine/analogs & derivatives , Cell Line, Tumor , Cell Proliferation , DNA Methylation , Decitabine , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , Transfection , ras Proteins/genetics , ras Proteins/metabolism
13.
Cancer Res ; 73(10): 3019-28, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23492365

ABSTRACT

Antiangiogenic strategies can be effective for cancer therapy, but like all therapies resistance poses a major clinical challenge. Hypoxia and nutrient starvation select for aggressive qualities that may render tumors resistant to antiangiogenic attack. Here, we show that hypoxia and nutrient starvation cooperate to drive tumor aggressiveness through epigenetic regulation of the histone demethylase JMJD1A (JHDM2A; KDM3A). In cancer cells rendered resistant to long-term hypoxia and nutrient starvation, we documented a stimulation of AKT phosphorylation, cell morphologic changes, cell migration, invasion, and anchorage-independent growth in culture. These qualities associated in vivo with increased angiogenesis and infiltration of macrophages into tumor tissues. Through expression microarray analysis, we identified a cluster of functional drivers such as VEGFA, FGF18, and JMJD1A, the latter which was upregulated in vitro under conditions of hypoxia and nutrient starvation and in vivo before activation of the angiogenic switch or the prerefractory phase of antiangiogenic therapy. JMJD1A inhibition suppressed tumor growth by downregulating angiogenesis and macrophage infiltration, by suppressing expression of FGF2, HGF, and ANG2. Notably, JMJD1A inhibition enhanced the antitumor effects of the anti-VEGF compound bevacizumab and the VEGFR/KDR inhibitor sunitinib. Our results form the foundation of a strategy to attack hypoxia- and nutrient starvation-resistant cancer cells as an approach to leverage antiangiogenic treatments and limit resistance to them.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Macrophages/drug effects , Neoplasms/drug therapy , Animals , Cell Hypoxia , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Jumonji Domain-Containing Histone Demethylases/physiology , Mice , Mice, Inbred C57BL , Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Pathologic/etiology , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Vascular Endothelial Growth Factor A/antagonists & inhibitors
15.
J Epidemiol ; 22(5): 384-94, 2012.
Article in English | MEDLINE | ID: mdl-22863985

ABSTRACT

Aberrant DNA methylation is associated with cancer development and progression. There are several types of specimens from which DNA methylation pattern can be measured and evaluated as an indicator of disease status (from normal biological process to pathologic condition) and even of pharmacologic response to therapy. Blood-based specimens such as cell-free circulating nucleic acid and DNA extracted from leukocytes in peripheral blood may be a potential source of noninvasive cancer biomarkers. In this article, we describe the characteristics of blood-based DNA methylation from different biological sources, detection methods, and the factors affecting DNA methylation. We provide a comprehensive literature review of blood-based DNA methylation as a cancer biomarker and focus on the study of DNA methylation using peripheral blood leukocytes. Although DNA methylation patterns measured in peripheral blood have great potential to be useful and informative biomarkers of cancer risk and prognosis, large systematic and unbiased prospective studies that consider biological plausibility and data analysis issues will be needed in order to develop a clinically feasible blood-based assay.


Subject(s)
Biomarkers, Tumor/metabolism , DNA Methylation , Leukocytes, Mononuclear/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Biomarkers, Tumor/blood , Humans , Molecular Epidemiology , Neoplasms/blood , Prognosis , Randomized Controlled Trials as Topic , Risk Factors
16.
Int J Cancer ; 131(11): 2596-603, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22447362

ABSTRACT

To determine whether or not the methylation status of blood leukocyte DNA can be used as a surrogate marker of the risk for cancer, we quantitatively determined the methylation levels of insulin-like growth factor 2 (IGF2) and TUSC3 in 299 gastric cancer cases, and 299 age- and gender-matched controls. The IGF2 methylation levels in blood leukocyte DNA of the cases were lower than those of the healthy controls and there was a significant trend of increasing gastric cancer risk with decreasing methylation level of IGF2. Patients with hypermethylated IGF2 in blood leukocyte DNA showed a significantly better survival rate than those with hypomethylated IGF2, indicating that the IGF2 methylation level in blood leukocyte DNA can be a possible marker not only of the risk for but also of the prognosis of gastric cancer. In contrast, the TUSC3 methylation level in blood leukocyte DNA was higher in the cases than in the healthy controls, but the difference was not significant. The past lifestyle and clinicopathological characteristics of the participants were analyzed for any relationship with the methylation level. With aging and smoking, methylation of IGF2 and TUSC3 decreased and increased in blood leukocyte DNA, respectively. These results indicate that the methylation level of IGF2 in blood leukocyte DNA may be used as an important surrogate marker of the risk for gastric cancer.


Subject(s)
DNA Methylation , DNA, Neoplasm/blood , DNA, Neoplasm/genetics , Insulin-Like Growth Factor II/genetics , Leukocytes/metabolism , Stomach Neoplasms/blood , Stomach Neoplasms/genetics , Aged , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Genetic Predisposition to Disease , Humans , Insulin-Like Growth Factor II/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Middle Aged , Prognosis , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
17.
PLoS One ; 7(3): e33608, 2012.
Article in English | MEDLINE | ID: mdl-22432036

ABSTRACT

BACKGROUND: To investigate the potential of serum miRNAs as biomarkers for early detection of gastric cancer (GC), a population-based study was conducted in Linqu, a high-risk area of GC in China. METHODOLOGY/PRINCIPAL FINDINGS: All subjects were selected from two large cohort studies. Differential miRNAs were identified in serum pools of GC and control using TaqMan low density array, and validated in individual from 82 pairs of GC and control, and 46 pairs of dysplasia and control by real-time quantitative reverse transcription-polymerase chain reaction. The temporal trends of identified serum miRNA expression were further explored in a retrospective study on 58 GC patients who had at least one pre-GC diagnosis serum sample based on the long-term follow-up population. The miRNA profiling results demonstrated that 16 miRNAs were markedly upregulated in GC patients compared to controls. Further validation identified a panel of three serum miRNAs (miR-221, miR-744, and miR-376c) as potential biomarkers for GC detection, and receiver operating characteristic (ROC) curve-based risk assessment analysis revealed that this panel could distinguish GCs from controls with 82.4% sensitivity and 58.8% specificity. MiR-221 and miR-376c demonstrated significantly positive correlation with poor differentiation of GC, and miR-221 displayed higher level in dysplasia than in control. Furthermore, the retrospective study revealed an increasing trend of these three miRNA levels during GC development (P for trend<0.05), and this panel could classify serum samples collected up to 5 years ahead of clinical GC diagnosis with 79.3% overall accuracy. CONCLUSIONS/SIGNIFICANCE: These data suggest that serum miR-221, miR-376c and miR-744 have strong potential as novel non-invasive biomarkers for early detection of GC.


Subject(s)
Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Early Detection of Cancer , MicroRNAs/blood , MicroRNAs/genetics , Stomach Neoplasms/blood , Stomach Neoplasms/genetics , Adult , Case-Control Studies , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Neoplasm Grading , ROC Curve , Reproducibility of Results , Retrospective Studies , Risk Assessment , Stomach Neoplasms/pathology , Time Factors
18.
Gut ; 61(3): 344-53, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21865403

ABSTRACT

BACKGROUND: Gastric cancer is the second most frequent cause of death from cancer in the world, diffuse-type gastric cancer (DGC) exhibiting a poor prognosis. Germline mutations of CDH1, encoding E-cadherin, have been reported in hereditary DGC, and genetic and/or epigenetic alterations of CDH1 are frequently detected in sporadic DGC. Genetic alterations of TP53 are also frequently found in DGC. To examine the synergistic effect of the loss of E-cadherin and p53 on gastric carcinogenesis, a mouse line was established in which E-cadherin and p53 are specifically inactivated in the stomach parietal cell lineage. METHODS: Atp4b-Cre mice were crossed with Cdh1(loxP/loxP) and Trp53(loxP/loxP) mice, and the gastric phenotype of Atp4b-Cre(+);Cdh1(loxP/loxP);Trp53(loxP/loxP) double conditional knockout (DCKO) mice was examined. RESULTS: Non-polarised E-cadherin-negative parietal cells and proton pump-negative atypical foci were observed in DCKO mice. Intramucosal cancers and invasive cancers composed of poorly differentiated carcinoma cells and signet ring cells, histologically very similar to those in humans, were found from 6 to 9 months, respectively. Fatal DGC developed at 100% penetrance within a year, frequently metastasised to lymph nodes, and had tumourigenic activity in immunodeficient mice. Gene expression profiles of DGC in DCKO mice also resembled those of human DGC, and mesenchymal markers and epithelial-mesenchymal transition-related genes were highly expressed in mouse DGC as in human DGC. CONCLUSION: This mouse line is the first genetically engineered mouse model of DGC and is very useful for clarifying the mechanism underlying gastric carcinogenesis, and provides a new approach to the treatment and prevention of DGC.


Subject(s)
Cadherins/physiology , Cell Transformation, Neoplastic/metabolism , Stomach Neoplasms/metabolism , Tumor Suppressor Protein p53/physiology , Animals , Cadherins/deficiency , Cell Polarity/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , DNA, Neoplasm/genetics , Disease Models, Animal , Gene Expression Profiling/methods , Genetic Predisposition to Disease , Immune Tolerance , Lymphatic Metastasis , Mice , Mice, Knockout , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis/methods , Parietal Cells, Gastric/metabolism , Parietal Cells, Gastric/pathology , Proton Pumps/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Tumor Suppressor Protein p53/deficiency
19.
Cancer Sci ; 102(11): 2007-13, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21848913

ABSTRACT

By behaving as molecular hubs, scaffold proteins can assemble a large number of signaling molecules and organize complicated intracellular signaling networks in time and space. Owing to their crucial role in mediating intracellular signaling related to tumor cell growth and migration, recent studies have highlighted the relevance of scaffold proteins in human cancers and indicated that interfering with their expression and/or their ability to bind effector proteins can inhibit cancer progression. Here, we show that receptor for activated C-kinase 1 (RACK1), a ubiquitously expressed scaffolding protein, plays a crucial regulatory role in tumor growth. Using an RNA silencing approach, we found that downregulation of RACK1 expression in HeLa and A673 tumor cells markedly suppressed the proliferation and invasion of these cells in vitro and tumor development in vivo. Consequently, we found that significant suppression of constitutive phosphorylation of Akt and MAPK by RACK1 silencing may contribute to the inhibition of tumor growth. Moreover, RACK1 silencing significantly attenuated tumor-associated angiogenesis by, at least in part, inhibiting the expression of two critical angiogenic factors, namely vascular endothelial growth factor-B and fibroblast growth factor 2. The results of the present study show that RACK1 is a potent enhancer of tumor growth and, thus, a potential anti-cancer therapeutic target.


Subject(s)
GTP-Binding Proteins/physiology , Neoplasm Proteins/physiology , RNA Interference , Receptors, Cell Surface/physiology , Animals , Apoptosis , Cell Division , Cell Line, Tumor , Down-Regulation , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness , Neovascularization, Pathologic , Phosphorylation , Protein Processing, Post-Translational , RNA, Small Interfering/pharmacology , Receptors for Activated C Kinase , Rhabdomyosarcoma/pathology , Signal Transduction , Tumor Stem Cell Assay , Vascular Endothelial Growth Factor B , Xenograft Model Antitumor Assays
20.
Cancer Sci ; 102(7): 1313-21, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21457403

ABSTRACT

CD133 is a universal marker of tissue stem/progenitor cells as well as cancer stem cells, but its physiological significance remains to be elucidated. Here we examined the relationship between expression of CD133 and features of gastric epithelial cells, and found that CD133-positive (CD133[+]) tumor cell lines formed well-differentiated tumors while CD133-negative (CD133[-]) lines formed poorly differentiated ones when subcutaneously injected into nude mice. We also found that CD133(+) and CD133(-) cell populations co-existed in some cell lines. FACS analysis showed that CD133(+) cells were mother cells because CD133(+) cells formed both CD133(+) and CD133(-) cells, but CD133(-) cells did not form CD133(+) cells. In these cell lines, CD133(+) cells formed well-differentiated tumors while CD133(-) cells formed poorly differentiated ones. In human gastric cancers, CD133 was exclusively expressed on the luminal surface membrane of gland-forming cells, and it was never found on poorly differentiated diffuse-type cells. Considering that poorly differentiated tumors often develop from well-differentiated tumors during tumor progression, these results suggest that loss of expression of CD133 might be related to gastric tumor progression. Microarray analysis showed that CD133(+) cells specifically expressed Sox17, a tumor suppressor in gastric carcinogenesis. Forced expression of SOX17 induced expression of CD133 in CD133(-) cells, and reduction of SOX17 caused by siRNA in CD133(+) cells induced a reduction in the level of CD133. These results indicate that Sox17 might be a key transcription factor controlling CD133 expression, and that it might also play a role in the control of gastric tumor progression.


Subject(s)
Antigens, CD/physiology , Glycoproteins/physiology , HMGB Proteins/physiology , Neoplastic Stem Cells/chemistry , Peptides/physiology , SOXF Transcription Factors/physiology , Stomach Neoplasms/pathology , AC133 Antigen , Animals , Antigens, CD/analysis , Female , Gastric Mucosa/pathology , Gene Expression Profiling , Glycoproteins/analysis , HMGB Proteins/analysis , Humans , Mice , Peptides/analysis , SOXF Transcription Factors/analysis , Stomach Neoplasms/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...