Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 197
Filter
1.
Pathobiology ; 78(1): 1-9, 2011.
Article in English | MEDLINE | ID: mdl-21474971

ABSTRACT

OBJECTIVE: The aim of this study is to clarify genome-wide DNA methylation profiles in renal tumors of various histological subtypes. METHODS: Bacterial artificial chromosome (BAC) array-based methylated CpG island amplification was performed using tissue samples of 17 patients with papillary renal cell carcinomas (RCCs), chromophobe RCCs and oncocytomas, and the results were compared with those from 51 patients with clear cell RCCs. RESULTS: Unsupervised hierarchical clustering analysis based on DNA methylation status clustered type 1 and type 2 papillary RCCs into different subclasses. Although chromophobe RCCs and oncocytomas were clustered into the same subclass, the DNA methylation status of 21 BAC clones was able to discriminate chromophobe RCCs from oncocytomas. The number of BAC clones showing DNA methylation alteration in non-tumorous renal tissue from patients with chromophobe RCCs and oncocytomas was smaller than that from patients with clear cell RCCs. Biphasic accumulation of DNA methylation alterations was observed in non-tumorous renal tissue from all 68 patients, and patients showing such alterations on more BAC clones had a poorer outcome than patients showing them on fewer BAC clones. CONCLUSIONS: DNA methylation profiles determining the histological subtypes of renal tumors developing in individual patients and/or patient outcome may be already established in non-tumorous renal tissue at the precancerous stage.


Subject(s)
Carcinoma, Renal Cell/genetics , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic/genetics , Kidney Neoplasms/genetics , Adenoma, Oxyphilic/genetics , Adenoma, Oxyphilic/pathology , Carcinoma, Renal Cell/pathology , Chromosomes, Artificial, Bacterial/genetics , Cluster Analysis , Female , Genome-Wide Association Study , Humans , Kidney Neoplasms/pathology , Male , Nephrectomy , Oligonucleotide Array Sequence Analysis , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Tissue Array Analysis
2.
Hepatol Res ; 41(3): 240-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21272165

ABSTRACT

AIM: Nuclear factor-κB (NF-κB) is a critical signaling mediator in inflammation, apoptosis resistance and oncogenesis. It has been reported that NF-κB is activated in several cancers, including hepatocellular carcinoma (HCC). Studies of genetic disruptions in mice also suggest that NF-κB plays critical roles in hepatocarcinogenesis. The aim of the present study is to characterize NF-κB activation and correlate it with the degree of malignancy in HCC. METHODS: To examine the correlation between the positivity of the nuclear p50 subunit and HCC recurrence, we analyzed immunostaining of the NF-κB p50 subunit in two groups of HCC samples with known prognosis and Akt phosphorylation status: 49 patients showing early recurrence within 6 months (group A) and 50 patients who were recurrence-free for at least for 3 years (group B). RESULTS: In group A, positive nuclear staining of p50 was shown in 18 cases (36.7%), whereas only one case (2.0%) in group B had positive nuclear staining of p50 (P = 2.48839 × 10(-5) ). This suggests a positive relationship between nuclear p50 and early recurrence and advanced HCC in humans. The presence of phosphorylated Akt correlated with nuclear staining of p50 in HCCs in group A (R(2) = 0.213, P < 0.001). CONCLUSION: Our results indicate that nuclear staining of p50 was clearly associated with early recurrent HCC, and the Akt pathway might play a role in NF-κB activation in a subset of early recurrent HCC.

3.
Hum Pathol ; 42(4): 558-67, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21239043

ABSTRACT

Dysadherin is a cancer-associated cell membrane glycoprotein that down-regulates E-cadherin and plays important roles in tumor progression and metastasis. Differentiated-type gastric carcinoma can be classified into 2 histologic subtypes according to the presence of poorly differentiated components: a mixed type (differentiated carcinoma with poorly differentiated components) and a pure type (purely differentiated-type adenocarcinoma). We studied the clinicopathologic features of 318 cases of differentiated-type gastric carcinoma with submucosal invasion and evaluated the immunohistochemical expression of dysadherin and E-cadherin. We also evaluated 46 cases of metastatic lymph nodes. Tumors with combined dysadherin-positive (≥50%) expression and E-cadherin-negative (<50%) expression had significantly higher proportions of the moderately differentiated type, deeper submucosal invasion, positivity of lymphatic permeation, and positivity of lymph node metastasis than tumors with other combinations of dysadherin and E-cadherin expression (P = .0009, P = .0015, P = .0273, and P = .0187, respectively). Moreover, the frequency of dysadherin-positive (≥50%) expression was higher in the mixed type (60.3%) than in the pure type (12.4%) (P < .0001), whereas the frequency of E-cadherin-negative (<50%) expression was higher in the mixed type (84.5%) than in the pure type (50.5%) (P < .0001). The frequency of dysadherin expression in the metastatic lymph nodes (80.4%) was significantly higher than that in the primary tumors (45.7%) (P = .001). Dysadherin-positive (≥50%) expression and E-cadherin-negative (<50%) expression may be correlated with the mixed type. Combined dysadherin-positive (≥50%) expression and E-cadherin-negative (<50%) expression may be valuable information for predicting aggressive tumor behavior.


Subject(s)
Cadherins/biosynthesis , Carcinoma/metabolism , Carcinoma/pathology , Gastric Mucosa/pathology , Membrane Glycoproteins/biosynthesis , Neoplasm Proteins/biosynthesis , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Aged , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Cell Differentiation , Female , Humans , Immunohistochemistry , Ion Channels , Male , Microfilament Proteins , Neoplasm Metastasis
4.
Gastroenterology ; 140(1): 310-21, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20955708

ABSTRACT

BACKGROUND & AIMS: Anti-tumor immunity changes over the course of tumor progression; it is not clear how or when the developing tumor overcomes immune surveillance. Intraductal papillary mucinous neoplasm (IPMN) is an intraepithelial precursor lesion of pancreatic cancer that progresses from adenoma to carcinoma. We investigated when and how the human anti-tumor immune reaction changes during pancreatic tumor development. METHODS: Using immunohistochemical analysis of cells isolated from patients with IPMN, the numbers of tumor-infiltrating lymphocytes and dendritic cells and the maturation state of dendritic cells in the regional lymph nodes were investigated during tumor progression. Gene expression profiles were compared among epithelial neoplastic cells at each stage of tumor development. Biological functions of the selected gene products were analyzed using syngeneic mouse models. RESULTS: The anti-tumor immune reaction changed from an immune response to immune tolerance between the stages of intraductal papillary mucinous adenoma (IPMA) and intraductal papillary mucinous carcinoma (IPMC). Chemokine (C-X-C motif) ligand 17 (CXCL17) and intercellular adhesion molecule 2 (ICAM2) were involved in immune surveillance during tumor development-their expression levels were up-regulated exclusively in IPMA and disappeared from IPMC. CXCL17 and ICAM2 induced infiltration and accumulation of the tumor epithelial layer by immature myeloid dendritic cells. This was followed by a cellular immune reaction and ICAM2 simultaneously promoted the susceptibility of the tumor cells to cytotoxic T-cell-mediated cytolysis. These processes had a synergistic effect to increase the anti-tumor immune response. CONCLUSIONS: Immune surveillance occurs during the early intraepithelial stages of human pancreatic carcinogenesis and is mediated by expression of CXCL17 and ICAM2.


Subject(s)
Adenocarcinoma, Mucinous/immunology , Adenocarcinoma, Papillary/immunology , Antigens, CD/immunology , Carcinoma in Situ/immunology , Carcinoma, Pancreatic Ductal/immunology , Cell Adhesion Molecules/immunology , Cell Transformation, Neoplastic/immunology , Chemokine CCL17/immunology , Immunologic Surveillance , Pancreatic Neoplasms/immunology , Precancerous Conditions/immunology , Abscess/immunology , Abscess/pathology , Abscess/surgery , Adenocarcinoma, Mucinous/pathology , Adenocarcinoma, Mucinous/surgery , Adenocarcinoma, Papillary/pathology , Adult , Aged , Animals , Carcinoma in Situ/pathology , Carcinoma, Pancreatic Ductal/pathology , Cell Transformation, Neoplastic/pathology , Dendritic Cells/immunology , Female , Gene Expression Profiling , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Male , Mice , Mice, Inbred BALB C , Middle Aged , Neoplasm Staging , Pancreatic Neoplasms/pathology , Pancreatic Pseudocyst/immunology , Pancreatic Pseudocyst/pathology , Pancreatitis, Chronic/immunology , Pancreatitis, Chronic/pathology , Precancerous Conditions/pathology , Retrospective Studies , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology
5.
Cancer Epidemiol Biomarkers Prev ; 20(1): 160-71, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21148121

ABSTRACT

BACKGROUND: Early detection is essential to improve the outcome of patients with pancreatic cancer. A noninvasive and cost-effective diagnostic test using plasma/serum biomarkers would facilitate the detection of pancreatic cancer at the early stage. METHODS: Using a novel combination of hollow fiber membrane-based low-molecular-weight protein enrichment and LC-MS-based quantitative shotgun proteomics, we compared the plasma proteome between 24 patients with pancreatic cancer and 21 healthy controls (training cohort). An identified biomarker candidate was then subjected to a large blinded independent validation (n = 237, validation cohort) using a high-density reverse-phase protein microarray. RESULTS: Among a total of 53,009 MS peaks, we identified a peptide derived from CXC chemokine ligand 7 (CXCL7) that was significantly reduced in pancreatic cancer patients, showing an area under curve (AUC) value of 0.84 and a P value of 0.00005 (Mann-Whitney U test). Reduction of the CXCL7 protein was consistently observed in pancreatic cancer patients including those with stage I and II disease in the validation cohort (P < 0.0001). The plasma level of CXCL7 was independent from that of CA19-9 (Pearson's r = 0.289), and combination with CXCL7 significantly improved the AUC value of CA19-9 to 0.961 (P = 0.002). CONCLUSIONS: We identified a significant decrease of the plasma CXCL7 level in patients with pancreatic cancer, and combination of CA19-9 with CXCL7 improved the discriminatory power of the former for pancreatic cancer. IMPACT: The present findings may provide a new diagnostic option for pancreatic cancer and facilitate early detection of the disease.


Subject(s)
Biomarkers, Tumor/blood , Pancreatic Neoplasms/blood , beta-Thromboglobulin/metabolism , Adult , Aged , CA-19-9 Antigen/blood , Case-Control Studies , Chromatography, Liquid , Female , Humans , Male , Mass Spectrometry , Middle Aged , Pancreatic Neoplasms/diagnosis , Prospective Studies , Reproducibility of Results , beta-Thromboglobulin/analysis
6.
Gastroenterology ; 140(3): 892-902, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21070779

ABSTRACT

BACKGROUND & AIMS: Two major types of gastric cancer, intestinal and diffuse, develop through distinct mechanisms; the diffuse type is considered to be more influenced by genetic factors, although the mechanism is unknown. Our previous genome-wide association study associated 3 single nucleotide polymorphisms (SNPs) with diffuse-type gastric cancer (DGC); 1 was a functional SNP (rs2294008) in prostate stem cell antigen (PSCA), but the loci of the other 2 were not investigated. METHODS: We performed high-density mapping to explore a linkage disequilibrium status of the 2 SNPs at chromosome 1q22. A DGC case-control study was conducted using DNA from 606 cases and 1264 controls (all Japanese individuals) and validated using DNA from Japanese (304 cases, 1465 controls) and Korean (452 cases, 372 controls) individuals. The effects of SNPs on function were analyzed by reporter assays and analyses of splice variants. RESULTS: A region of a strong linkage disequilibrium with the 2 SNPs contained mucin 1 (MUC1) and other 4 genes and SNPs significantly associated with DGC (rs2070803: P = 4.33 × 10(-13); odds ratio [OR], 1.71 by meta-analysis of the studies on the 3 panels) but not with intestinal-type gastric cancer. Functional studies demonstrated that rs4072037 (P = 1.43 × 10(-11); OR, 1.66 by meta-analysis) in MUC1 affects promoter activity and determines the major splicing variants of MUC1 in the gastric epithelium. Individuals that carry both SNPs rs2294008 in PSCA and rs4072037 in MUC1 have a high risk for developing DGC (OR, 8.38). CONCLUSIONS: MUC1 is the second major DGC susceptibility gene identified. The SNPs rs2070803 and rs4072037 in MUC1 might be used to identify individuals at risk for this type of gastric cancer.


Subject(s)
Chromosomes, Human, Pair 1 , Mucin-1/genetics , Polymorphism, Single Nucleotide , Stomach Neoplasms/genetics , Asian People/genetics , Case-Control Studies , Cell Line, Tumor , Exons , Female , Genetic Predisposition to Disease , Genome-Wide Association Study , Haplotypes , Humans , Japan/epidemiology , Linkage Disequilibrium , Logistic Models , Male , Middle Aged , Mucin-1/metabolism , Odds Ratio , Phenotype , Promoter Regions, Genetic , Republic of Korea/epidemiology , Risk Assessment , Risk Factors , Stomach Neoplasms/ethnology , Stomach Neoplasms/pathology , Transfection
7.
Proteomics Clin Appl ; 4(5): 560-7, 2010 May.
Article in English | MEDLINE | ID: mdl-21137073

ABSTRACT

PURPOSE: We aimed to identify novel chemotherapy responsiveness biomarkers for osteosarcoma (OS) by investigating the global protein expression profile of 12 biopsy samples from OS patients. EXPERIMENTAL DESIGN: Six patients were classified as good responders and six as poor responders, according to the Huvos grading system. The protein expression profiles obtained by 2-D DIGE consisted of 2250 protein spots. RESULTS: Among them, we identified 55 protein spots whose intensity was significantly different (Bonferroni adjusted p-value<0.01) between the two patient groups. Mass spectrometric protein identification demonstrated that the 55 spots corresponded to 38 distinct gene products including peroxiredoxin 2 (PRDX 2). Use of a specific antibody against PRDX 2 confirmed the differential expression of PRDX 2 between good and poor responders, while PRDX 2 levels as measured by Western blotting correlated highly with their corresponding 2-D DIGE values. The predictive value of PRDX 2 expression was further confirmed by examining an additional four OS cases using Western blotting. CONCLUSIONS AND CLINICAL RELEVANCE: These results establish PRDX 2 as a candidate for chemotherapy responsiveness marker in OS. Measuring PRDX 2 in biopsy samples before treatment may contribute to more effective management of OS.


Subject(s)
Bone Neoplasms/drug therapy , Osteosarcoma/drug therapy , Peroxiredoxins/metabolism , Adolescent , Adult , Biomarkers, Tumor/metabolism , Bone Neoplasms/metabolism , Child , Female , Gene Expression Profiling , Humans , Male , Osteosarcoma/metabolism , Prognosis , Proteomics/methods
8.
Cancer Res ; 70(22): 9095-105, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-21062981

ABSTRACT

In multicellular organisms, adaptive responses to oxidative stress are regulated by NF-E2-related factor 2 (NRF2), a master transcription factor of antioxidant genes and phase II detoxifying enzymes. Aberrant activation of NRF2 by either loss-of-function mutations in the Keap1 gene or gain-of-function mutations in the Nrf2 gene occurs in a wide range of human cancers, but details of the biological consequences of NRF2 activation in the cancer cells remain unclear. Here, we report that mutant NRF2 induces epithelial cell proliferation, anchorage-independent growth, and tumorigenicity and metastasis in vivo. Genome-wide gene expression profiling revealed that mutant NRF2 affects diverse molecular pathways including the mammalian target of rapamycin (mTOR) pathway. Mutant NRF2 upregulates RagD, a small G-protein activator of the mTOR pathway, which was also overexpressed in primary lung cancer. Consistently, Nrf2-mutated lung cancer cells were sensitive to mTOR pathway inhibitors (rapamycin and NVP-BEZ235) in both in vitro and an in vivo xenograft model. The gene expression signature associated with mutant NRF2 was a marker of poor prognosis in patients with carcinoma of the head and neck region and lung. These results show that oncogenic Nrf2 mutation induces dependence on the mTOR pathway during carcinogenesis. Our findings offer a rationale to target NRF2 as an anticancer strategy, and they suggest NRF2 activation as a novel biomarker for personalized molecular therapies or prognostic assessment.


Subject(s)
Gene Expression Profiling/methods , Mutation , NF-E2-Related Factor 2/genetics , Signal Transduction/genetics , TOR Serine-Threonine Kinases/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , HEK293 Cells , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Nude , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous , Tumor Burden/genetics
9.
Histol Histopathol ; 25(10): 1257-67, 2010 10.
Article in English | MEDLINE | ID: mdl-20712010

ABSTRACT

Dysadherin, a cancer associated cell membrane glycoprotein, has been reported to downregulate E-cadherin. Aberrant expression of E-cadherin has been associated with the development of metastases in patients with cancer. Even though the expression of dysadherin and E-cadherin has been studied in primary non-small cell lung carcinoma, little is known about its expression at the distant metastases sites. We investigate by immunohistochemistry the relationship between E-cadherin and dysadherin in 111 cases of primary lung carcinomas (53 squamous cell carcinomas, 21 adenocarcinomas, 13 large cell carcinomas, and 24 small cell carcinomas), and their distant metastases. The intensity, the expression pattern and the percentage of neoplastic cell staining were recorded and the results were correlated with clinicopathological findings of the subjects. Dysadherin immunostain was expressed in 61 (54.95%) of the cases, and increased dysadherin expression was significantly correlated with tumour size (p=0.003), distant metastases (p=0.0034), and metastasis size (p=0.0008). Reduced E-cadherin expression was noted in 46 (41.45%) of the cases, and was correlated with high-grade tumour (p=0.02), infiltrative growth pattern (p=0.042), and advanced stage (p=0.032). Although the correlation between the expression of dysadherin and E-cadherin was not significant, a group of patients showed reduced E-cadherin expression with dysadherin overexpression. In lung carcinomas dysadherin expression seems to reflect tumour aggressiveness and may be considered a positive marker of poor prognosis when considered alone or/and in combination with down-regulation of E-cadherin.


Subject(s)
Adenocarcinoma/chemistry , Biomarkers, Tumor/analysis , Cadherins/analysis , Carcinoma, Large Cell/chemistry , Carcinoma, Non-Small-Cell Lung/chemistry , Carcinoma, Small Cell/chemistry , Carcinoma, Squamous Cell/chemistry , Lung Neoplasms/chemistry , Membrane Glycoproteins/analysis , Neoplasm Proteins/analysis , Adenocarcinoma/secondary , Aged , Aged, 80 and over , Antigens, CD , Carcinoma, Large Cell/secondary , Carcinoma, Non-Small-Cell Lung/secondary , Carcinoma, Small Cell/secondary , Carcinoma, Squamous Cell/secondary , Chi-Square Distribution , Female , Greece , Humans , Immunohistochemistry , Ion Channels , Lung Neoplasms/pathology , Male , Microfilament Proteins , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Prognosis
10.
J Biol Chem ; 285(34): 26289-94, 2010 Aug 20.
Article in English | MEDLINE | ID: mdl-20566648

ABSTRACT

Wnt signaling pathways play important roles in various stages of developmental events and several aspects of adult homeostasis. Aberrant activation of Wnt signaling has also been associated with several types of cancer. We have recently identified Traf2- and Nck-interacting kinase (TNIK) as a novel activator of Wnt signaling through a comprehensive proteomic approach in human colorectal cancer cell lines. TNIK is an activating kinase for T-cell factor-4 (TCF4) and essential for the beta-catenin-TCF4 transactivation and colorectal cancer growth. Here, we report the essential role of TNIK in Wnt signaling during Xenopus development. We found that Xenopus TNIK (XTNIK) was expressed maternally and that the functional knockdown of XTNIK by catalytically inactive XTNIK (K54R) or antisense morpholino oligonucleotides resulted in significant malformations with a complete loss of head and axis structures. XTNIK enhanced beta-catenin-induced axis duplication and the expression of beta-catenin-TCF target genes, whereas knockdown of XTNIK inhibited it. XTNIK was recruited to the promoter region of beta-catenin-TCF target genes in a beta-catenin-dependent manner. These results demonstrate that XTNIK is an essential factor for the transcriptional activity of the beta-catenin-TCF complex and dorsal axis determination in Xenopus embryos.


Subject(s)
Body Patterning , Protein Serine-Threonine Kinases/physiology , Signal Transduction/physiology , Wnt Proteins/metabolism , Xenopus Proteins/physiology , Abnormalities, Multiple/etiology , Animals , Embryo, Nonmammalian , Germinal Center Kinases , Transcription, Genetic , Xenopus/embryology , beta Catenin
11.
Am J Clin Pathol ; 134(1): 71-9, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20551269

ABSTRACT

To assess the expression of a cancer-associated fibroblasts (CAFs) marker as an indicator of prognosis, we raised anti-protein gene product 9.5 (PGP9.5) monoclonal antibody against cultured fibroblasts. PGP9.5 expression in cultured normal fibroblasts was increased by transforming growth factor beta stimulation, indicating the phenotypic alteration to activated fibroblast. We immunohistochemically evaluated PGP9.5 expression with the CAFs of 110 colorectal cancer cases under T3 stage. PGP9.5 immunoreactivity in 30% or more of CAFs was defined as high PGP9.5 expression, and the other cases were considered as having low PGP9.5 expression. Patients with high PGP9.5 expression (42.7%) had significantly shorter survival and a higher incidence of recurrence than the low PGP9.5 expression group (P = .002 and P < .001, respectively). Multivariate analysis indicated PGP9.5 expression as an independent prognostic factor for overall and recurrence-free survival partly as well as lymph node metastasis. These results indicate that PGP9.5 expression in CAFs is a helpful finding to represent the overall biologic behavior of advanced colorectal cancer.


Subject(s)
Adenocarcinoma/metabolism , Antibodies, Monoclonal/metabolism , Biomarkers, Tumor/metabolism , Colorectal Neoplasms/metabolism , Fibroblasts/metabolism , Ubiquitin Thiolesterase/metabolism , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Biomarkers, Tumor/chemistry , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Electrophoresis, Polyacrylamide Gel , Female , Humans , Immunoenzyme Techniques , Japan/epidemiology , Male , Mice , Mice, Inbred BALB C , Middle Aged , Neoplasm Staging , Prognosis , Proportional Hazards Models , Survival Rate , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/pharmacology , Tumor Cells, Cultured , Ubiquitin Thiolesterase/chemistry , Ubiquitin Thiolesterase/immunology
12.
Cancer Res ; 70(12): 5024-33, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20530691

ABSTRACT

T-cell factor-4 (TCF4) is a transcription factor essential for maintaining the undifferentiated status and self-renewal of intestinal epithelial cells. It has therefore been considered that constitutive activation of TCF4 by aberrant Wnt signaling is a major force driving colorectal carcinogenesis. We previously identified Traf2- and Nck-interacting kinase (TNIK) as one of the proteins that interact with TCF4 in colorectal cancer cells, but its functional significance has not been elucidated. Here, we report that TNIK is an activating kinase for TCF4 and essential for colorectal cancer growth. TNIK, but not its catalytically inactive mutant, phosphorylated the conserved serine 154 residue of TCF4. Small interfering RNA targeting TNIK inhibited the proliferation of colorectal cancer cells and the growth of tumors produced by injecting colorectal cancer cells s.c. into immunodeficient mice. The growth inhibition was abolished by restoring the catalytic domain of TNIK, thus confirming that its enzyme activity is essential for the maintenance of colorectal cancer growth. Several ATP-competing kinase inhibitors have been applied to cancer treatment and have shown significant activity. Our findings suggest TNIK as a feasible target for pharmacologic intervention to ablate aberrant Wnt signaling in colorectal cancer.


Subject(s)
Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , TNF Receptor-Associated Factor 2/metabolism , Wnt Proteins/metabolism , Adenoviridae/genetics , Animals , Blotting, Western , Cell Proliferation , Chromatography, Liquid , Colony-Forming Units Assay , Colorectal Neoplasms/genetics , Female , Germinal Center Kinases , Humans , Immunoblotting , Immunoenzyme Techniques , Immunoprecipitation , Luciferases/metabolism , Mass Spectrometry , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorylation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , TNF Receptor-Associated Factor 2/genetics , Tumor Cells, Cultured , Two-Hybrid System Techniques , Wnt Proteins/genetics , beta Catenin/genetics , beta Catenin/metabolism
13.
Clin Cancer Res ; 16(9): 2518-28, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20388846

ABSTRACT

PURPOSE: The outcome of patients with advanced hepatocellular carcinoma (HCC) has remained unsatisfactory. Patients with HCC suffer from chronic hepatitis or liver cirrhosis, and their reserve liver function is often limited. EXPERIMENTAL DESIGN: To develop new therapeutic agents that act specifically on HCC but interfere only minimally with residual liver function, we searched for genes that were upregulated in 20 cases of HCC [namely, discovery sets 1 (n = 10) and 2 (n = 10)] in comparison with corresponding nontumorous liver and a panel representing normal organs using high-density microarrays capable of detecting all exons in the human genome. RESULTS: Eleven transcripts whose expression was significantly increased in HCC were subjected to siRNA-based secondary screening of genes required for HCC cell proliferation as well as quantitative reverse transcription-PCR analysis [validation sets 1 (n = 20) and 2 (n = 44)] and immunohistochemistry (n = 19). We finally extracted four genes, AKR1B10, HCAP-G, RRM2, and TPX2, as candidate therapeutic targets for HCC. siRNA-mediated knockdown of these candidate genes inhibited the proliferation of HCC cells and the growth of HCC xenografts transplanted into immunodeficient mice. CONCLUSIONS: The four genes we identified were highly expressed in HCC, and HCC cells are highly dependent on these genes for proliferation. Although many important genes must have been overlooked, the selected genes were biologically relevant. The combination of genome-wide expression and functional screening described here is a rapid and comprehensive approach that could be applied in the identification of therapeutic targets in any type of human malignancy.


Subject(s)
Carcinoma, Hepatocellular/genetics , Genome, Human/genetics , Genome-Wide Association Study/methods , Liver Neoplasms/genetics , Aged , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation , Cluster Analysis , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease/genetics , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/pathology , Liver Neoplasms, Experimental/prevention & control , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Oligonucleotide Array Sequence Analysis , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous
14.
Mol Cancer Ther ; 9(3): 535-44, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20159990

ABSTRACT

Pulmonary metastasis is the most significant prognostic determinant for osteosarcoma, but methods for its prediction and treatment have not been established. Using oligonucleotide microarrays, we compared the global gene expression of biopsy samples between seven osteosarcoma patients who developed pulmonary metastasis within 4 years after neoadjuvant chemotherapy and curative resection, and 12 patients who did not relapse. We identified argininosuccinate synthetase (ASS) as a gene differentially expressed with the highest statistical significance (Welch's t test, P = 2.2 x 10(-5)). Immunohistochemical analysis of an independent cohort of 62 osteosarcoma cases confirmed that reduced expression of ASS protein was significantly correlated with the development of pulmonary metastasis after surgery (log-rank test, P < 0.05). Cox regression analysis revealed that ASS was the sole significant predictive factor (P = 0.039; hazard ratio, 0.319; 95% confidence interval, 0.108-0.945). ASS is one of the enzymes required for the production of a nonessential amino acid, arginine. We showed that osteosarcoma cells lacking ASS expression were auxotrophic for arginine and underwent G(0)-G(1) arrest in arginine-free medium, suggesting that an arginine deprivation therapy could be effective in patients with osteosarcoma. Recently, phase I and II clinical trials in patients with melanoma and hepatocellular carcinoma have shown the safety and efficacy of plasma arginine depletion by stabilized arginine deiminase. Our data indicate that in patients with osteosarcoma, reduced expression of ASS is not only a novel predictive biomarker for the development of metastasis, but also a potential target for pharmacologic intervention.


Subject(s)
Argininosuccinate Synthase/genetics , Bone Neoplasms/diagnosis , Lung Neoplasms/diagnosis , Lung Neoplasms/secondary , Osteosarcoma/diagnosis , Adolescent , Adult , Argininosuccinate Synthase/metabolism , Argininosuccinate Synthase/physiology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/physiology , Bone Neoplasms/genetics , Bone Neoplasms/mortality , Bone Neoplasms/pathology , Child , Down-Regulation/physiology , Female , Gene Expression Profiling , Gene Expression Regulation, Enzymologic/physiology , Gene Expression Regulation, Neoplastic/physiology , Humans , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Male , Oligonucleotide Array Sequence Analysis , Osteosarcoma/genetics , Osteosarcoma/mortality , Osteosarcoma/pathology , Prognosis , Tumor Cells, Cultured , Young Adult
15.
Cancer Sci ; 101(4): 882-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20088962

ABSTRACT

The aim of this study was to establish new biliary tract carcinoma (BTC) cell lines and identify predictive biomarkers for the potential effectiveness of gemcitabine therapy. Surgical specimens of BTC were transplanted directly into immunodeficient mice to establish xenografts, then subjected to in vitro cell culture. The gemcitabine sensitivity of each cell line was determined and compared with the genome-wide gene expression profile. A new predictive biomarker candidate was validated using an additional cohort of gemcitabine-treated BTC cases. From 55 BTC cases, we established 19 xenografts and six new cell lines. Based on their gemcitabine sensitivity, 10 BTC cell lines (including six new and four publicly available ones) were clearly categorized into two groups, and MAGEH1 mRNA expression in the tumor cells showed a significant negative correlation with their sensitivity to gemcitabine. Immunohistochemically, MAGEH1 protein was detected in three (50%) out of six sensitive cell lines, and four (100%) out of four resistant cell lines. In the validation cohort of gemcitabine-treated recurrence cases, patients were categorized into "effective" and "non-effective" groups according to the RECIST guidelines for assessment of chemotherapeutic effects. MAGEH1 protein expression was detected in two (40%) out of five "effective" cases and all four (100%) "non-effective" cases. We have established a new BTC bioresource that covers a wide range of biological features, including drug sensitivity, and is linked with clinical information. Negative expression of MAGEH1 protein serves as a potential predictive marker for the effectiveness of gemcitabine therapy in BTC.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Biliary Tract Neoplasms/drug therapy , Deoxycytidine/analogs & derivatives , Animals , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/metabolism , Biliary Tract Neoplasms/metabolism , Deoxycytidine/therapeutic use , Female , Gene Expression Profiling , Humans , Mice , Mice, SCID , Microtubule-Associated Proteins , Neoplasm Proteins , Specific Pathogen-Free Organisms , Gemcitabine
16.
Mol Cell Proteomics ; 9(4): 695-704, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20061307

ABSTRACT

Although gemcitabine monotherapy is the standard treatment for advanced pancreatic cancer, patient outcome varies significantly, and a considerable number do not benefit adequately. We therefore searched for new biomarkers predictive of overall patient survival. Using LC-MS, we compared the base-line plasma proteome between 29 representative patients with advanced pancreatic cancer who died within 100 days and 31 patients who survived for more than 400 days after receiving at least two cycles of the same gemcitabine monotherapy. Identified biomarker candidates were then challenged in a larger cohort of 304 patients treated with the same protocol using reverse-phase protein microarray. Among a total of 45,277 peptide peaks, we identified 637 peaks whose intensities differed significantly between the two groups (p < 0.001, Welch's t test). Two MS peaks with the highest statistical significance (p = 2.6 x 10(-4) and p = 5.0 x 10(-4)) were revealed to be derived from alpha(1)-antitrypsin and alpha(1)-antichymotrypsin, respectively. The levels of alpha(1)-antitrypsin (p = 8.9 x 10(-8)) and alpha(1)-antichymotrypsin (p = 0.001) were significantly correlated with the overall survival of the 304 patients. We selected alpha(1)-antitrypsin (p = 0.0001), leukocyte count (p = 0.066), alkaline phosphatase (p = 8.3 x 10(-8)), and performance status (p = 0.003) using multivariate Cox regression analysis and constructed a scoring system (nomogram) that was able to identify a group of high risk patients having a short median survival time of 150 days (95% confidence interval, 123-187 days; p = 2.0 x 10(-15), log rank test). The accuracy of this model for prognostication was internally validated and showed good calibration and discrimination with a bootstrap-corrected concordance index of 0.672. In conclusion, an increased level of alpha(1)-antitrypsin is a biomarker that predicts short overall survival of patients with advanced pancreatic cancer receiving gemcitabine monotherapy. Although an external validation study will be necessary, the current model may be useful for identifying patients unsuitable for the standardized therapy.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/mortality , Deoxycytidine/analogs & derivatives , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/mortality , Aged , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/isolation & purification , Biomarkers, Tumor/metabolism , Calibration , Carcinoma, Pancreatic Ductal/diagnosis , Carcinoma, Pancreatic Ductal/metabolism , Deoxycytidine/therapeutic use , Early Detection of Cancer/methods , Early Detection of Cancer/standards , Female , Humans , Male , Middle Aged , Neoplasm Proteins/analysis , Neoplasm Proteins/isolation & purification , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/metabolism , Prognosis , Retrospective Studies , Survival Analysis , Gemcitabine
17.
Cancer Sci ; 101(1): 231-40, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19775289

ABSTRACT

To clarify genome-wide DNA methylation profiles during multistage urothelial carcinogenesis, bacterial artificial chromosome (BAC) array-based methylated CpG island amplification (BAMCA) was performed in 18 normal urothelia obtained from patients without urothelial carcinomas (UCs) (C), 17 noncancerous urothelia obtained from patients with UCs (N), and 40 UCs. DNA hypo- and hypermethylation on multiple BAC clones was observed even in N compared to C. Principal component analysis revealed progressive DNA methylation alterations from C to N, and to UCs. DNA methylation profiles in N obtained from patients with invasive UCs were inherited by the invasive UCs themselves, that is DNA methylation alterations in N were correlated with the development of more malignant UCs. The combination of DNA methylation status on 83 BAC clones selected by Wilcoxon test was able to completely discriminate N from C, and diagnose N as having a high risk of carcinogenesis, with 100% sensitivity and specificity. The combination of DNA methylation status on 20 BAC clones selected by Wilcoxon test was able to completely discriminate patients who suffered from recurrence after surgery from patients who did not. The combination of DNA methylation status for 11 BAC clones selected by Wilcoxon test was able to completely discriminate patients with UCs of the renal pelvis or ureter who suffered from intravesical metachronous UC development from patients who did not. Genome-wide alterations of DNA methylation may participate in urothelial carcinogenesis from the precancerous stage to UC, and DNA methylation profiling may provide optimal indicators for carcinogenetic risk estimation and prognostication.


Subject(s)
DNA Methylation , Precancerous Conditions/genetics , Urinary Bladder Neoplasms/genetics , Aged , Aged, 80 and over , Chromosomes, Artificial, Bacterial , CpG Islands , Female , Humans , Male , Middle Aged , Precancerous Conditions/pathology , Urinary Bladder Neoplasms/pathology
18.
Jpn J Clin Oncol ; 40(1): 60-72, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19815537

ABSTRACT

OBJECTIVE: The clinical course of gastrointestinal stromal tumor (GIST) spans a wide spectrum from a curable disorder to a highly malignant disease that leads to metastasis and death. To develop prognostic modalities for GIST patients, we developed a mouse monoclonal antibody against pfetin, the prognostic value of which has been previously reported. METHODS: The reactivity of the monoclonal antibody against pfetin was examined by western blotting and immunohistochemistry. RESULTS: Western blotting demonstrated that the monoclonal antibody was specific to pfetin. The immunohistochemical study demonstrated that the 5-year disease-free survival rate was 93.2% and 94.5% for GIST patients with pfetin-positive tumors and 70.0% and 80.7% for those with pfetin-negative tumors in the 159 cases from the National Cancer Center Hospital (P < 0.0001) and in the 100 cases from Niigata University Medical and Dental Hospital (P < 0.0001), respectively. Uni- and multivariate analyses revealed that pfetin expression was a powerful prognostic factor among the clinico-pathological parameters examined. CONCLUSIONS: These results establish pfetin as a practical prognostic marker for GIST patients after surgery. Pfetin may also present a novel therapeutic target to prevent recurrence of GIST.


Subject(s)
Antibodies, Monoclonal , Biomarkers, Tumor/blood , Gastrointestinal Stromal Tumors/diagnosis , Proteins/analysis , Adult , Aged , Aged, 80 and over , Animals , Biomarkers, Tumor/chemistry , Biomarkers, Tumor/immunology , Blotting, Western , Cell Line, Tumor , Electrophoresis, Polyacrylamide Gel , Female , Gastrointestinal Stromal Tumors/immunology , Gastrointestinal Stromal Tumors/physiopathology , Humans , Immunohistochemistry , Male , Mice , Middle Aged , Predictive Value of Tests , Prognosis , Proteins/chemistry , Proteins/genetics
19.
Pancreas ; 38(7): e200-6, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19893451

ABSTRACT

OBJECTIVES: Pancreatic cancer is one of the most intractable of cancers. However, the comprehensive view of somatic mutations in this tumor is far from clear. The tyrosine kinase (TK) gene family, which encodes important regulators of various signal transduction pathways, is one of the most frequently altered gene families in human cancer. METHODS: To clarify the somatic mutation profile of TKs in pancreatic cancer, we performed a systematic screening of mutations in the kinase domains of all human TK genes (636 exons of 90 genes in total) in 11 pancreatic cancer cell lines and 29 microdissected primary tumors. RESULTS: We identified 15 nonsynonymous alterations that included 9 DNA alterations in cell lines and 6 somatic mutations in primary tumors. In particular, we identified the previously reported pathogenic mutation of NTRK3 in a KRAS/BRAF wild-type tumor and 2 somatic mutations in the Src family of kinases (YES1 and LYN) that would be expected to cause structural changes. CONCLUSIONS: Our genome-wide resequencing approach revealed novel oncogenic pathways in pancreatic cancers.


Subject(s)
Mutation , Pancreatic Neoplasms/genetics , Protein-Tyrosine Kinases/genetics , Sequence Analysis, DNA/methods , Adult , Aged , Cell Line, Tumor , Female , Genetic Predisposition to Disease/genetics , Genetic Testing , Genome, Human/genetics , Genome-Wide Association Study , Humans , Male , Middle Aged , Multigene Family , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-yes/genetics , Proto-Oncogene Proteins p21(ras) , Receptor, trkC/genetics , ras Proteins/genetics , src-Family Kinases/genetics
20.
Cancer Sci ; 100(12): 2268-74, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19725836

ABSTRACT

Osteosarcoma (OS) is the most frequent primary malignant bone tumor of children and young adults. Although the introduction of combined neoadjuvant chemotherapy has markedly improved survival, the outcome of OS patients with distant metastasis and/or poor response to chemotherapy is still unsatisfactory. Therefore there is a need to develop new therapeutic agents that suppress OS cell proliferation with higher efficacy. The protein kinases are a family of genes that play critical roles in various signaling pathways. Some cancer cells show addiction to constitutive activation of certain signaling pathways for proliferation and survival. To identify new drug targets for OS, we screened a panel of small interfering RNAs (siRNAs) that target 691 genes encoding human protein kinases and related proteins. We found that different constructs of siRNA specifically targeting polo-like 1 kinase (PLK1) significantly caused mitotic cell cycle arrest and subsequent apoptotic cell death in a variety of OS cell lines. siRNA targeting PLK1 also suppressed the growth of OS xenografts established in immunodeficient mice. Recently, phase I clinical trials of PLK1 chemical inhibitors have been reported. Our results indicate that PLK1 is a promising molecular target for pharmacologic intervention in OS.


Subject(s)
Bone Neoplasms/therapy , Cell Cycle Proteins/antagonists & inhibitors , Osteosarcoma/therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , RNA, Small Interfering/therapeutic use , Animals , Bone Neoplasms/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , Humans , Male , Mice , Mice, Inbred BALB C , Osteosarcoma/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Polo-Like Kinase 1
SELECTION OF CITATIONS
SEARCH DETAIL
...