Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Cancer Genomics Proteomics ; 15(5): 413-420, 2018.
Article in English | MEDLINE | ID: mdl-30194082

ABSTRACT

BACKGROUND/AIM: Overall survival for the high-risk group of neuroblastoma (NB) patients still remains at 40-50%, necessitating the establishment of a curable treatment. LIM domain only 1 (LMO1) gene encoding a transcriptional regulator is an NB-susceptibility gene with a tumor-promoting activity. Previously we conducted chromatin immunoprecipitation and DNA sequencing analyses on NB cell lines and identified 3 protein-coding genes regulated by LMO1. In this study, we extended our analyses to capture microRNA genes directly or indirectly regulated by LMO1. MATERIALS AND METHODS: Using microarrays, we conducted a comparative gene expression analysis on an NB cell line SK-N-SH; between the cells with and without LMO1 suppression. RESULTS: Overall, 18 microRNAs were identified to be indirectly down-regulated by LMO1 including 7 microRNAs of the let-7 family, whose cell proliferation inhibitory activity was observed. CONCLUSION: Target genes of the LMO1-regulated microRNAs and their relevant pathways may be a potential therapeutic target.


Subject(s)
DNA-Binding Proteins/genetics , LIM Domain Proteins/genetics , MicroRNAs/genetics , Neuroblastoma/genetics , Transcription Factors/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation/genetics , Humans , Microarray Analysis/methods , Neuroblastoma/pathology
2.
Cancer Genomics Proteomics ; 15(3): 165-174, 2018.
Article in English | MEDLINE | ID: mdl-29695398

ABSTRACT

BACKGROUND/AIM: Overall survival for the high-risk group of neuroblastoma (NB) remains at 40-50%. An integrative genomics study revealed that LIM domain only 1 (LMO1) encoding a transcriptional regulator to be an NB-susceptibility gene with a tumor-promoting activity, that needs to be revealed. MATERIALS AND METHODS: We conducted chromatin immunoprecipitation and DNA sequencing analyses and cell proliferation assays on two NB cell lines. RESULTS: We identified three genes regulated by LMO1 in the cells, LIM and senescent cell antigen-like domains 1 (LIMS1), Ras suppressor protein 1 (RSU1) and relaxin 2 (RLN2). LIMS1 and RSU1 encode proteins functioning with integrin-linked kinase (ILK), and inhibition of LIMS1, ILK or RLN2 by shRNA reduced cell proliferation of the NB cells, which was also suppressed with an ILK inhibiting compound Cpd 22. CONCLUSION: The downstream of LMO1-regulatory cascade includes a tumor-promoting LIMS1/ILK pathway, which has a potential to be a novel therapeutic target.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Chromatin Immunoprecipitation/methods , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , High-Throughput Nucleotide Sequencing/methods , LIM Domain Proteins/genetics , Neuroblastoma/genetics , Protein Serine-Threonine Kinases/genetics , Transcription Factors/genetics , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Cell Proliferation , DNA-Binding Proteins/antagonists & inhibitors , Humans , LIM Domain Proteins/antagonists & inhibitors , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , Protein Serine-Threonine Kinases/antagonists & inhibitors , RNA, Small Interfering/genetics , Signal Transduction , Transcription Factors/antagonists & inhibitors , Tumor Cells, Cultured
3.
Oncol Lett ; 15(3): 3081-3084, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29435040

ABSTRACT

Prostate stem cell antigen (PSCA) is a glycosylphosphatidylinositol (GPI)-anchored cell surface protein and exhibits an organ-dependent expression pattern in cancer. PSCA is upregulated in prostate cancer and downregulated in gastric cancer. PSCA is expressed in a variety of human organs. Although certain studies previously demonstrated its expression in the mammalian and avian brain, its expression in the human brain has not been thoroughly elucidated. Additionally, it was previously reported that PSCA is weakly expressed in the astrocytes of the normal human brain but aberrantly expressed in glioma, suggesting that PSCA is a promising target of glioma therapy and prostate cancer therapy. The current study identified PSCA expression in the neural and choroid plexus cells of the normal human brain by immunohistochemistry. In brain tumors, PSCA was expressed in medulloblastoma and glioma, and its expression was also observed in papilloma and papillary carcinoma of the choroid plexus, ependymoma and meningioma. The results suggest that PSCA may have a tumor-promoting function in brain tumors and is a potential target for their therapy. However, its expression in normal neuronal and choroid plexus cells implies that a PSCA-targeted therapy may lead to certain adverse phenomena.

4.
Anticancer Res ; 35(5): 2619-25, 2015 May.
Article in English | MEDLINE | ID: mdl-25964537

ABSTRACT

BACKGROUND/AIM: In previous work, we found that prostate stem cell antigen (PSCA) gene, encoding a glycosylphosphatidylinositol-anchored protein, is a presumable tumor suppressor in gastric cancer and gallbladder cancer (GBC). The introduction of PSCA cDNA into GBC cell lines significantly suppressed tumorigenecity of cells in mice. The PSCA protein is thought to be involved in some form of intracellular signaling that remains to be elucidated. MATERIALS AND METHODS: Using microarrays, we conducted gene-expression profiling on tumors generated by a GBC cell line TGBC-1TKB, with and without expression of PSCA, which was implanted into mice. Genes whose expression was down-regulated by PSCA were selected, and their down-regulation was confirmed by real-time PCR. RESULTS: We identified several immune-related genes down-regulated by PSCA, including interleukin 8 (IL8), IL1 receptor antagonist (IL1RN) and S100 calcium-binding proteins A8 (S100A8) and A9 (S100A9). CONCLUSION: PSCA signaling may suppress tumor growth in vivo by modulating immunological characteristics of GBC cells.


Subject(s)
Antigens, Neoplasm/genetics , Gallbladder Neoplasms/genetics , Glycosylphosphatidylinositols/biosynthesis , Neoplasm Proteins/genetics , Animals , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/immunology , Calgranulin A/biosynthesis , Calgranulin A/immunology , Calgranulin B/biosynthesis , Calgranulin B/immunology , Cell Line, Tumor , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Gallbladder Neoplasms/immunology , Gallbladder Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Glycosylphosphatidylinositols/genetics , Glycosylphosphatidylinositols/immunology , Humans , Interleukin 1 Receptor Antagonist Protein/biosynthesis , Interleukin 1 Receptor Antagonist Protein/immunology , Interleukin-8/biosynthesis , Interleukin-8/immunology , Mice , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/immunology
5.
BMC Cancer ; 15: 439, 2015 May 29.
Article in English | MEDLINE | ID: mdl-26016667

ABSTRACT

BACKGROUND: Gastric cancer (GC) is one of the major malignant diseases worldwide, especially in Asia, and Japan and Korea have the highest incidence in the world. Because most of the cases that are refractory to therapies die due to peritoneal dissemination (PD) of the cancer cells, controlling PD is important for patient survival. GSDMB is a member of the gasdermin gene family. Because GSDMB is expressed in many types of cancer, including GC, it is likely that the gene contains a regulatory region that is utilized for therapy of occult PD through cancer cell-specific expression of cytotoxic genes. METHODS: We performed reporter assays to identify the regulatory region for the cancer cell-specific expression. We also constructed a lentiviral therapeutic vector that expresses herpes simplex virus thymidine kinase (HSVtk) in a GC cell-specific manner, and tested it in a mouse model of PD. RESULTS: We identified the regulatory region at +496 to +989 from the GSDMB transcription start site and designated it as a GSDMB enhancer. The lentiviral therapeutic vector suppressed proliferation of a GC cell line, 60As6, in vitro in the presence of ganciclovir, and intraperitoneal administration of the vector prolonged the survival term of mice that were intraperitoneally inoculated with 60As6 one week prior to the administration. CONCLUSIONS: The GSDMB-driven HSVtk expression vector had a therapeutic effect on the occult PD model mice. This strategy can potentially be used to treat GC patients with PD.


Subject(s)
Genetic Therapy , Neoplasm Proteins/genetics , Peritoneal Neoplasms/therapy , Stomach Neoplasms/therapy , Thymidine Kinase/genetics , Animals , Genetic Vectors/therapeutic use , Humans , Mice , Peritoneal Cavity/pathology , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/pathology , Peritoneum/pathology , Primary Cell Culture , Promoter Regions, Genetic/genetics , Regulatory Sequences, Nucleic Acid/genetics , Simplexvirus/enzymology , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Thymidine Kinase/biosynthesis
6.
Genes Cells ; 20(5): 382-91, 2015 May.
Article in English | MEDLINE | ID: mdl-25727947

ABSTRACT

Previous genomewide association studies identified prostate stem cell antigen (PSCA) as a gastric cancer (GC) susceptibility gene and showed an association between GC and the T allele of the single nucleotide polymorphism rs2294008 (C/T) in this gene. The protein product of this gene inhibits cell growth, and the T allele significantly suppresses the transcriptional activity of the -3.2 kb PSCA upstream region. However, the mechanism remains unknown. In this study, we conducted reporter assays using the PSCA upstream region containing the C allele and identified the region from -200 to +38 bp of the transcription initiation site of the gene as a critical region of the -3.2 kb PSCA upstream region. We found that introducing the T allele at rs2294008 generated a consensus binding sequence for the Polycomb group transcription factor Yin Yang 1 (YY1) and that disruption of the consensus sequence restored the transcriptional activity to the -3.2 kb PSCA upstream region. These findings imply that the T allele significantly suppresses PSCA expression in vivo by recruiting YY1 to its promoter, which eventually predisposes gastric epithelial cells to GC development.


Subject(s)
Alleles , Antigens, Neoplasm/genetics , Gallbladder Neoplasms/genetics , Gallbladder Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/genetics , Promoter Regions, Genetic , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , YY1 Transcription Factor/metabolism , Base Sequence , Binding Sites , Consensus Sequence , GPI-Linked Proteins/genetics , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Gene Order , Humans , Molecular Sequence Data , Protein Binding , Transcription, Genetic
7.
Int J Mol Sci ; 15(5): 7958-73, 2014 May 07.
Article in English | MEDLINE | ID: mdl-24810688

ABSTRACT

Gastric cancer (GC) is one of the major malignant diseases worldwide, especially in Asia. It is classified into intestinal and diffuse types. While the intestinal-type GC (IGC) is almost certainly caused by Helicobacter pylori (HP) infection, its role in the diffuse-type GC (DGC) appears limited. Recently, genome-wide association studies (GWAS) on Japanese and Chinese populations identified chromosome 1q22 as a GC susceptibility locus which harbors mucin 1 gene (MUC1) encoding a cell membrane-bound mucin protein. MUC1 has been known as an oncogene with an anti-apoptotic function in cancer cells; however, in normal gastric mucosa, it is anticipated that the mucin 1 protein has a role in protecting gastric epithelial cells from a variety of external insults which cause inflammation and carcinogenesis. HP infection is the most definite insult leading to GC, and a protective function of mucin 1 protein has been suggested by studies on Muc1 knocked-out mice.


Subject(s)
Mucin-1/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Stomach/pathology , Animals , Gastric Mucosa/metabolism , Gene Knockdown Techniques , Genome-Wide Association Study , Helicobacter/immunology , Helicobacter/isolation & purification , Helicobacter Infections/complications , Helicobacter Infections/immunology , Humans , Inflammation/complications , Inflammation/immunology , Inflammation/microbiology , Mice , Mucin-1/immunology , Polymorphism, Single Nucleotide , Stomach/immunology , Stomach/microbiology , Stomach Neoplasms/immunology , Stomach Neoplasms/microbiology
8.
J Carcinog ; 12: 4, 2013.
Article in English | MEDLINE | ID: mdl-23599686

ABSTRACT

BACKGROUND: Prostate stem cell antigen (PSCA), an organ-dependent tumor suppressor, is down regulated in gallbladder cancer (GBC). It is anticipated that the missense allele C of the single nucleotide polymorphism (SNP) rs2294008 (T/C) in the translation initiation codon of the gene affects the gene's biological function and has some influence on GBC susceptibility. We examined the biological effect of the C allele on the function of the gene and the relation between the C allele and GBC susceptibility. MATERIALS AND METHODS: Functional analysis of the SNP was conducted by introducing PSCA cDNA harboring the allele to a GBC cell line TGBC- 1TKB and performing colony formation assays in vitro and tumor formation assays in mice. The effect on transcriptional regulation was assessed by reporter assays. The association study was conducted on 44 Japanese GBC cases and 173 controls. RESULTS: The PSCA cDNA harboring the C allele showed lower cell growth inhibition activity (20% reduction) than that with the T allele. Concordantly, when injected into subcutaneous tissues of mice, the GBC cell line stably expressing the cDNA with the C allele formed tumors of almost the same size as that of the control cells, but the cell line expressing the cDNA with the T allele showed slower growth. The upstream DNA fragment harboring the C allele had more transcriptional activity than that with the T allele. The C allele showed positive correlation to GBC but no statistical significant odds ratio (OR = 1.77, 95% confidence interval 0.85-3.70, P value = 0.127 in dominant model). CONCLUSIONS: The missense allele was shown to have a biological effect, attenuating antitumor activities of PSCA, and consequently it may be a potential risk for GBC development. An association study in a larger sample size may reveal a significant association between the allele and GBC.

9.
Cancer Sci ; 104(1): 1-8, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23057512

ABSTRACT

Gastric cancer (GC) is one of the major malignant diseases worldwide, especially in Asia, where Japan and Korea have the highest incidence in the world. Gastric cancer is classified into intestinal and diffuse types. While the former is almost absolutely caused by Helicobacter pylori infection as the initial insult, the latter seems to include cases in which the role of infection is limited, if any, and a contribution of genetic factors is anticipated. Previously, we performed a genome-wide association study (GWAS) on diffuse-type GC by using single nucleotide polymorphisms (SNP) catalogued for Japanese population (JSNP), and identified a prostate stem cell antigen (PSCA) gene encoding a glycosylphosphatidylinositol-anchored cell surface antigen as a GC susceptibility gene. From the second candidate locus identified using the GWAS, 1q22, we found the Mucin 1 (MUC1) gene encoding a cell membrane-bound mucin protein as another gene related to diffuse-type GC. A two-allele analysis based on risk genotypes of the two genes revealed approximately 95% of Japanese population have at least one of the two risk genotypes, and approximately 56% of the population have both risk genotypes. The two-SNP genotype might offer ample room to further stratify a high GC risk subpopulation in Japan and Asia by adding another genetic and/or non-genetic factor. Recently, a GWAS on the Chinese population disclosed an additional three GC susceptibility loci: 3q13.31, 5p13.1 and 10q23.


Subject(s)
Antigens, Neoplasm/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , Mucin-1/genetics , Neoplasm Proteins/genetics , Stomach Neoplasms/genetics , Asia/epidemiology , China/epidemiology , GPI-Linked Proteins/genetics , Genotype , Glycosylphosphatidylinositols/genetics , Humans , Japan/epidemiology , Polymorphism, Single Nucleotide , Stomach Neoplasms/epidemiology
10.
Anat Cell Biol ; 45(3): 149-54, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23094202

ABSTRACT

Prostate stem cell antigen (PSCA) is a glycosylphosphatidylinositol-anchored cell surface antigen with an organ-dependent expression pattern in cancers; e.g., up-regulated in prostate cancer and down-regulated in gastric cancer. Previously it was reported that PSCA is not expressed in the normal pancreas but aberrantly expressed in pancreatic cancer. In this present study, we identified PSCA expression in islets of the pancreas by immunohistochemistry, which was co-localized with four islet-cell markers: insulin, glucagon, somatostatin and pancreatic polypeptide. In our investigation of the transcription start site of PSCA, we found a non-coding splicing variant of PSCA as well as authentic PSCA transcripts in mRNA samples from a normal pancreas. Both the transcripts were also identified in several pancreatic cancer cell lines. We previously reported that PSCA expression is correlated to the methylation status of the enhancer region in gastric and gallbladder cancer cell lines but not in pancreatic cancer cell lines, suggesting that PSCA expression is regulated in a diff erent mode in pancreatic cancer from that in gastric and gallbladder cancers.

11.
13.
Genes Chromosomes Cancer ; 51(1): 30-41, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21936014

ABSTRACT

Gallbladder cancer (GBC) is relatively rare but has a high mortality rate. One candidate molecule which might be involved in GBC development is prostate stem cell antigen (PSCA), a glycosylphosphatidylinositol-anchored cell surface antigen with a tissue-specific pattern of expression in the epithelium of several organs, such as the prostate, stomach, bladder, and gallbladder. It is up-regulated in a number of cancers including prostate, urinary bladder, and pancreatic cancers, while it is down-regulated in esophageal and gastric cancers, suggesting that PSCA has an oncogenic activity in the former but a tumor suppressor activity in the latter. However, the precise function of PSCA and the regulatory mechanism for its expression in normal and cancer cells are yet to be determined. In this study, immunohistochemical analyses with a specific antibody revealed that PSCA is down-regulated in non-neoplastic gallbladder lesions such as cholesterolosis, cholecystolithiasis, and cholecystitis (9/17; 53%), and also in adenocarcinoma (40/44; 91%), a common neoplasm in gallbladder. Analyses of the DNA methylation status in the GBC cell lines by bisulfite-Pyrosequencing and a reporter assay for the PSCA promoter activity suggested that the down-regulation is explained, at least partly, by DNA methylation. Moreover, colony formation assay revealed that PSCA has cell-proliferation inhibition activity in the GBC cell lines, which was also observed in vivo. These lines of in vivo and in vitro evidence suggest that PSCA is acting as a tumor suppressor in GBC development.


Subject(s)
Antigens, Neoplasm/genetics , Cell Transformation, Neoplastic/genetics , Down-Regulation/genetics , Gallbladder Neoplasms/genetics , Genes, Tumor Suppressor , Neoplasm Proteins/genetics , Cell Line, Tumor , Cell Proliferation , Cholecystitis/genetics , DNA Methylation , Epithelium/metabolism , GPI-Linked Proteins/genetics , Gallbladder/metabolism , Humans , Male , Promoter Regions, Genetic
14.
Nature ; 469(7329): 216-20, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-21124317

ABSTRACT

Neuroblastoma is a childhood cancer of the sympathetic nervous system that accounts for approximately 10% of all paediatric oncology deaths. To identify genetic risk factors for neuroblastoma, we performed a genome-wide association study (GWAS) on 2,251 patients and 6,097 control subjects of European ancestry from four case series. Here we report a significant association within LIM domain only 1 (LMO1) at 11p15.4 (rs110419, combined P = 5.2 × 10(-16), odds ratio of risk allele = 1.34 (95% confidence interval 1.25-1.44)). The signal was enriched in the subset of patients with the most aggressive form of the disease. LMO1 encodes a cysteine-rich transcriptional regulator, and its paralogues (LMO2, LMO3 and LMO4) have each been previously implicated in cancer. In parallel, we analysed genome-wide DNA copy number alterations in 701 primary tumours. We found that the LMO1 locus was aberrant in 12.4% through a duplication event, and that this event was associated with more advanced disease (P < 0.0001) and survival (P = 0.041). The germline single nucleotide polymorphism (SNP) risk alleles and somatic copy number gains were associated with increased LMO1 expression in neuroblastoma cell lines and primary tumours, consistent with a gain-of-function role in tumorigenesis. Short hairpin RNA (shRNA)-mediated depletion of LMO1 inhibited growth of neuroblastoma cells with high LMO1 expression, whereas forced expression of LMO1 in neuroblastoma cells with low LMO1 expression enhanced proliferation. These data show that common polymorphisms at the LMO1 locus are strongly associated with susceptibility to developing neuroblastoma, but also may influence the likelihood of further somatic alterations at this locus, leading to malignant progression.


Subject(s)
DNA-Binding Proteins/genetics , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Neuroblastoma/genetics , Oncogenes/genetics , Transcription Factors/genetics , Alleles , Cell Line, Tumor , Cell Proliferation , Chromosomes, Human, Pair 11/genetics , DNA Copy Number Variations/genetics , Disease Progression , Europe/ethnology , Gene Duplication/genetics , Gene Expression Regulation, Neoplastic/genetics , Genome, Human/genetics , Genomics , Genotype , Humans , LIM Domain Proteins , Neuroblastoma/pathology , Odds Ratio , Phenotype , Polymorphism, Single Nucleotide/genetics , Survival Rate
15.
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
16.
Cancer Sci ; 101(7): 1582-9, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20507324

ABSTRACT

The power of an SNP-based genome-wide association study (GWAS) was first demonstrated in Japan using the JSNP database and is currently a major strategy adopted around the world for a number of common diseases including cancers. The hypothesis-free strategy can lead us to a novel hypothesis for carcinogenesis and may contribute to identifying a high risk group for research and, in the future, practice of personalized prevention. We performed a GWAS on diffuse-type gastric cancer and identified a significant association with SNPs in the PSCA (prostate stem cell antigen) gene. The association was validated by a Korean gastric case-control analysis. The PSCA protein is expressed predominantly in the stem cell/precursor-rich region of the gastric epithelium, which is considered as the origin of diffuse-type gastric cancer, and showed tumor suppressor-like characteristics. Individuals with a low PSCA promoter activity are susceptible to diffuse-type gastric cancer. By contrast, the polymorphism does not significantly predispose to intestinal-type gastric cancer, congruous to the hypothesis of the two distinct carcinogenesis pathways for the two major types of gastric cancer. In addition to publication on a specific gene, the sharing of GWAS data through a database on the web is expected to accelerate validation and discovery by other investigators. GeMDBJ (Genome Medicine Database of Japan), started in 2005 in Japan, is one of such attempts. Moreover, the advent of "next generation" sequencers may herald a new era in which the poorly explored domains of the genetic architecture of disease susceptibility may be unveiled.


Subject(s)
Genome-Wide Association Study , Neoplasms/genetics , Polymorphism, Single Nucleotide , Stomach Neoplasms/genetics , Adenocarcinoma/genetics , Antigens, Neoplasm/genetics , GPI-Linked Proteins , Genes, Dominant , Genetic Predisposition to Disease , Humans , Japan , Korea/epidemiology , Linkage Disequilibrium , Membrane Glycoproteins/genetics , Models, Genetic , Neoplasm Proteins/genetics , Risk Assessment , Risk Factors , Stomach Neoplasms/epidemiology
17.
Clin Cancer Res ; 16(14): 3533-8, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20501618

ABSTRACT

Prostate stem cell antigen (PSCA) is a glycosylphosphatidylinositol (GPI)-anchored cell surface protein. Although PSCA is thought to be involved in intracellular signaling, much remains unknown about its physiological function and regulatory mechanism in normal and cancer cells. It is up-regulated in several major cancers including prostate, bladder, and pancreatic cancers. The expression of PSCA is positively correlated with advanced clinical stage and metastasis in prostate cancers and is also associated with malignant progression of premalignant prostate lesions. Therefore, PSCA has been proposed as a biomarker of diagnosis and prognosis, as well as a target of therapy for these cancers. In addition, PSCA has also shown clinical potential in immunotherapy as a prostate-specific antigen, which, when presented by dendritic cells, may elicit strong tumor-specific immunity. In contrast, PSCA is down-regulated in esophageal and gastric cancer and may have a tumor-suppressing function in the gastric epithelium. Recent exciting findings that genetic variations of PSCA conferred increased risks of gastric cancer and bladder cancer have opened up a new avenue of research about the pathological function of PSCA. PSCA seems to be a Jekyll and Hyde molecule that plays differential roles, tumor promoting or suppressing, depending on the cellular context.


Subject(s)
Antigens, Neoplasm/metabolism , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/genetics , Prostatic Neoplasms/genetics , Urinary Bladder Neoplasms/genetics , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Biomarkers, Tumor/metabolism , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , GPI-Linked Proteins/metabolism , Humans , Male , Models, Biological , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/metabolism , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/metabolism , Urinary Bladder Neoplasms/diagnosis , Urinary Bladder Neoplasms/metabolism
18.
Nat Genet ; 41(9): 991-5, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19648920

ABSTRACT

We conducted a genome-wide association study on 969 bladder cancer cases and 957 controls from Texas. For fast-track validation, we evaluated 60 SNPs in three additional US populations and validated the top SNP in nine European populations. A missense variant (rs2294008) in the PSCA gene showed consistent association with bladder cancer in US and European populations. Combining all subjects (6,667 cases, 39,590 controls), the overall P-value was 2.14 x 10(-10) and the allelic odds ratio was 1.15 (95% confidence interval 1.10-1.20). rs2294008 alters the start codon and is predicted to cause truncation of nine amino acids from the N-terminal signal sequence of the primary PSCA translation product. In vitro reporter gene assay showed that the variant allele significantly reduced promoter activity. Resequencing of the PSCA genomic region showed that rs2294008 is the only common missense SNP in PSCA. Our data identify rs2294008 as a new bladder cancer susceptibility locus.


Subject(s)
Antigens, Neoplasm/genetics , Genetic Predisposition to Disease , Genetic Variation , Membrane Glycoproteins/genetics , Neoplasm Proteins/genetics , Urinary Bladder Neoplasms/genetics , Alleles , Antigens, Neoplasm/immunology , Case-Control Studies , Chromosome Mapping , Chromosomes, Human, Pair 8 , GPI-Linked Proteins , Gene Frequency , Genes, Reporter , Genome-Wide Association Study , Haplotypes , Heterozygote , Homozygote , Humans , Linkage Disequilibrium , Logistic Models , Male , Multivariate Analysis , Mutation, Missense , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Reproducibility of Results , Urinary Bladder Neoplasms/immunology
19.
Nihon Rinsho ; 67(6): 1227-32, 2009 Jun.
Article in Japanese | MEDLINE | ID: mdl-19507519

ABSTRACT

A genome-wide association study on diffuse-type gastric cancer identified the PSCA gene, whose exact function in the gastric epithelium has been unknown. Immunohistochemical analysis revealed that PSCA is expressed in the region in which gastric epithelial stem cells and precursors are considered to reside. Moreover, the PSCA was downregulated in gastric cancer tissues, and its product showed a cell-growth inhibition activity in vitro. We also identified a functional SNP, the risk allele of which suppressed a promoter activity of the PSCA gene. These findings suggest that the PSCA may contribute to carcinogenesis through the cell-growth regulation of the gastric epithelial precursors. The summary statistics of the 2-stage genome scan is available from a database, GeMDBJ (http://gemdbj.nibio.go.jp/).


Subject(s)
Genome-Wide Association Study , Stomach Neoplasms/genetics , Antigens, Neoplasm , Databases, Genetic , GPI-Linked Proteins , Humans , Membrane Glycoproteins/genetics , Neoplasm Proteins/genetics
20.
Genes Chromosomes Cancer ; 48(3): 261-71, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19051310

ABSTRACT

Gasdermin (GSDM or GSDMA), expressed in the upper gastrointestinal tract but frequently silenced in gastric cancers (GCs), regulates apoptosis of the gastric epithelium. It has three human homologs, GSDMB, GSDMC, and GSDMD (GSDM family) and they are considered to be involved in the regulation of epithelial apoptosis but not yet known. We investigated the expression pattern of the family genes in the upper gastrointestinal epithelium and cancers. Reverse transcriptase-polymerase chain reaction revealed that, unlike GSDMA expressed in differentiated cells, GSDMB is expressed in proliferating cells and GSDMD in differentiating cells. GSDMC, meanwhile, is expressed in both differentiating and differentiated cells. Colony formation assay showed that GSDMB, closely related to GSDMA, has no cell-growth inhibition activity in gastric cancer cells, and that GSDMC and GSDMD, respectively, exhibit the activity with different strengths from that of GSDMA. Expression analyses of the four family genes in esophageal and GCs suggested that GSDMC and GSDMD as well as GSDMA are tumor suppressors and that GSDMB, which was amplified and overexpressed in some GCs, could be an oncogene. The results of the expression analysis and colony formation assay suggest that each family gene may have a distinct function in the upper gastrointestinal epithelium.


Subject(s)
Epithelial Cells/metabolism , Esophageal Neoplasms/genetics , Esophagus/metabolism , Gastric Mucosa/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Stomach Neoplasms/genetics , Amino Acid Sequence , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epithelial Cells/cytology , Epithelial Cells/pathology , Esophageal Neoplasms/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Phosphate-Binding Proteins , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...