Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
1.
Leukemia ; 32(2): 419-428, 2018 02.
Article in English | MEDLINE | ID: mdl-28720764

ABSTRACT

Both proto-oncogenic and tumor-suppressive functions have been reported for enhancer of zeste homolog 2 (EZH2). To investigate the effects of its inactivation, a mutant EZH2 lacking its catalytic domain was prepared (EZH2-dSET). In a mouse bone marrow transplant model, EZH2-dSET expression in bone marrow cells induced a myelodysplastic syndrome (MDS)-like disease in transplanted mice. Analysis of these mice identified Abcg2 as a direct target of EZH2. Intriguingly, Abcg2 expression alone induced the same disease in the transplanted mice, where stemness genes were enriched. Interestingly, ABCG2 expression is specifically high in MDS patients. The present results indicate that ABCG2 de-repression induced by EZH2 mutations have crucial roles in MDS pathogenesis.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Animals , Disease Models, Animal , Mice , Mutation/genetics
2.
Oncogene ; 36(45): 6262-6271, 2017 11 09.
Article in English | MEDLINE | ID: mdl-28692045

ABSTRACT

Epigenetic gene regulation linked to oncogenic pathways is an important focus of cancer research. KDM3A, a histone H3 lysine 9 (H3K9) demethylase, is known to have a pro-tumorigenic function. Here, we showed that KDM3A contributes to liver tumor formation through the phosphatidylinositol 3-kinase (PI3K) pathway, which is often activated in hepatocellular carcinoma. Loss of Kdm3a attenuated tumor formation in Pik3ca transgenic (Tg) mouse livers. Transcriptome analysis of pre-cancerous liver tissues revealed that the expression of activator protein 1 (AP-1) target genes was induced by PI3K activation, but blunted upon Kdm3a ablation. Particularly, the expression of Cd44, a liver cancer stem marker, was regulated by AP-1 in a Kdm3a-dependent manner. We identified Cd44-positive hepatocytes with epithelial-mesenchymal transition-related expression profiles in the Pik3ca Tg liver and confirmed their in vivo tumorigenic capacity. Notably, the number and tumor-initiating capacity of Cd44-positive hepatocytes were governed by Kdm3a. As a mechanism in Kdm3a-dependent AP-1 transcription, Kdm3a recruited c-Jun to the AP-1 binding sites of Cd44, Mmp7 and Pdgfrb without affecting c-Jun expression. Moreover, Brg1, a component of the SWI/SNF chromatin remodeling complex, interacted with c-Jun in a Kdm3a-dependent manner and was bound to the AP-1 binding site of these genes. Finally, KDM3A and c-JUN were co-expressed in 33% of human premalignant lesions with PI3K activation. Our data suggest a critical role for KDM3A in the PI3K/AP-1 oncogenic axis and propose a novel strategy for inhibition of KDM3A against liver tumor development under PI3K pathway activation.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Liver Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Transcription Factor AP-1/metabolism , Animals , Carcinogenesis , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Epigenesis, Genetic , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/enzymology , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Male , Mice , Mice, Knockout , Mice, Transgenic , Phosphorylation , Signal Transduction
3.
Oncogene ; 36(35): 4963-4974, 2017 08 31.
Article in English | MEDLINE | ID: mdl-28459464

ABSTRACT

Bone morphogenetic protein (BMP) signaling exerts antitumor activities in glioblastoma; however, its precise mechanisms remain to be elucidated. Here, we demonstrated that the BMP type I receptor ALK-2 (encoded by the ACVR1 gene) has crucial roles in apoptosis induction of patient-derived glioma-initiating cells (GICs), TGS-01 and TGS-04. We also characterized a BMP target gene, Distal-less homeobox 2 (DLX2), and found that DLX2 promoted apoptosis and neural differentiation of GICs. The tumor-suppressive effects of ALK-2 and DLX2 were further confirmed in a mouse orthotopic transplantation model. Interestingly, valproic acid (VPA), an anti-epileptic compound, induced BMP2, BMP4, ACVR1 and DLX2 mRNA expression with a concomitant increase in phosphorylation of Smad1/5. Consistently, we showed that treatment with VPA induced apoptosis of GICs, whereas silencing of ALK-2 or DLX2 expression partially suppressed it. Our study thus reveals BMP-mediated inhibitory mechanisms for glioblastoma, which explains, at least in part, the therapeutic effects of VPA.


Subject(s)
Activin Receptors, Type I/metabolism , Bone Morphogenetic Proteins/metabolism , Brain Neoplasms/metabolism , Glioma/metabolism , Homeodomain Proteins/metabolism , Neoplastic Stem Cells/metabolism , Transcription Factors/metabolism , Valproic Acid/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Differentiation/physiology , Female , Glioma/drug therapy , Glioma/pathology , HEK293 Cells , Heterografts , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Phosphorylation , Signal Transduction/drug effects , Transfection
4.
Oncogene ; 35(49): 6350-6358, 2016 12 08.
Article in English | MEDLINE | ID: mdl-27270436

ABSTRACT

Androgen receptor (AR) functions as a ligand-dependent transcription factor to regulate its downstream signaling for prostate cancer progression. AR complex formation by multiple transcription factors is important for enhancer activity and transcriptional regulation. However, the significance of such collaborative transcription factors has not been fully understood. In this study, we show that Oct1, an AR collaborative factor, coordinates genome-wide AR signaling for prostate cancer growth. Using global analysis by chromatin immunoprecipitation sequencing (ChIP-seq), we found that Oct1 is recruited to AR-binding enhancer/promoter regions and facilitates androgen signaling. Moreover, a major target of AR/Oct1 complex, acyl-CoA synthetase 3 (ACSL3), contributes to tumor growth in nude mice, and its high expression is associated with poor prognosis in prostate cancer patients. Next, we examined the therapeutic effects of pyrrole-imidazole polyamides that target the Oct1-binding sequence identified in the center of the ACSL3 AR-binding site. We observed that treatment with Oct1 polyamide severely blocked the Oct1 binding at the ACSL3 enhancer responsible for its transcriptional activity and ACSL3 induction. In addition, Oct1 polyamides suppressed castration-resistant tumor growth and specifically repressed global Oct1 chromatin association and androgen signaling in prostate cancer cells, with few nonspecific effects on basal promoter activity. Thus, targeting Oct1 binding could be a novel therapeutic strategy for AR-activated castration-resistant prostate cancer.


Subject(s)
Androgen Receptor Antagonists/pharmacology , Nylons/pharmacology , Octamer Transcription Factor-1/genetics , Prostatic Neoplasms, Castration-Resistant/genetics , Receptors, Androgen/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Coenzyme A Ligases/genetics , Coenzyme A Ligases/metabolism , Genomics , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , Male , Mice , Mice, Nude , Molecular Targeted Therapy , Nylons/chemistry , Octamer Transcription Factor-1/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Pyrroles/chemistry , Pyrroles/pharmacology , Receptors, Androgen/genetics , Signal Transduction , Transfection
5.
Blood Cancer J ; 4: e264, 2014 Dec 12.
Article in English | MEDLINE | ID: mdl-25501021

ABSTRACT

TET2 (Ten Eleven Translocation 2) is a dioxygenase that converts methylcytosine (mC) to hydroxymethylcytosine (hmC). TET2 loss-of-function mutations are highly frequent in subtypes of T-cell lymphoma that harbor follicular helper T (Tfh)-cell-like features, such as angioimmunoblastic T-cell lymphoma (30-83%) or peripheral T-cell lymphoma, not otherwise specified (10-49%), as well as myeloid malignancies. Here, we show that middle-aged Tet2 knockdown (Tet2(gt/gt)) mice exhibit Tfh-like cell overproduction in the spleen compared with control mice. The Tet2 knockdown mice eventually develop T-cell lymphoma with Tfh-like features after a long latency (median 67 weeks). Transcriptome analysis revealed that these lymphoma cells had Tfh-like gene expression patterns when compared with splenic CD4-positive cells of wild-type mice. The lymphoma cells showed lower hmC densities around the transcription start site (TSS) and higher mC densities at the regions of the TSS, gene body and CpG islands. These epigenetic changes, seen in Tet2 insufficiency-triggered lymphoma, possibly contributed to predated outgrowth of Tfh-like cells and subsequent lymphomagenesis. The mouse model described here suggests that TET2 mutations play a major role in the development of T-cell lymphoma with Tfh-like features in humans.


Subject(s)
Cell Transformation, Neoplastic/metabolism , DNA-Binding Proteins/biosynthesis , Lymphoma, Follicular/metabolism , Lymphoma, T-Cell/metabolism , Neoplasms, Experimental/metabolism , Proto-Oncogene Proteins/biosynthesis , T-Lymphocytes, Helper-Inducer/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , DNA-Binding Proteins/genetics , Dioxygenases , Gene Knockdown Techniques , Humans , Lymphoma, Follicular/genetics , Lymphoma, Follicular/pathology , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/pathology , Mice , Mice, Transgenic , Mutation , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Proto-Oncogene Proteins/genetics , T-Lymphocytes, Helper-Inducer/pathology
6.
Leukemia ; 28(2): 241-7, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24220272

ABSTRACT

High-throughput DNA sequencing significantly contributed to diagnosis and prognostication in patients with myelodysplastic syndromes (MDS). We determined the biological and prognostic significance of genetic aberrations in MDS. In total, 944 patients with various MDS subtypes were screened for known/putative mutations/deletions in 104 genes using targeted deep sequencing and array-based genomic hybridization. In total, 845/944 patients (89.5%) harbored at least one mutation (median, 3 per patient; range, 0-12). Forty-seven genes were significantly mutated with TET2, SF3B1, ASXL1, SRSF2, DNMT3A, and RUNX1 mutated in >10% of cases. Many mutations were associated with higher risk groups and/or blast elevation. Survival was investigated in 875 patients. By univariate analysis, 25/48 genes (resulting from 47 genes tested significantly plus PRPF8) affected survival (P<0.05). The status of 14 genes combined with conventional factors revealed a novel prognostic model ('Model-1') separating patients into four risk groups ('low', 'intermediate', 'high', 'very high risk') with 3-year survival of 95.2, 69.3, 32.8, and 5.3% (P<0.001). Subsequently, a 'gene-only model' ('Model-2') was constructed based on 14 genes also yielding four significant risk groups (P<0.001). Both models were reproducible in the validation cohort (n=175 patients; P<0.001 each). Thus, large-scale genetic and molecular profiling of multiple target genes is invaluable for subclassification and prognostication in MDS patients.


Subject(s)
Myelodysplastic Syndromes/genetics , Adult , Aged , Aged, 80 and over , Female , Gene Frequency , Genetic Association Studies , Genetic Markers , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged , Mutation , Mutation Rate , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/mortality , Polymorphism, Single Nucleotide , Prognosis , Proportional Hazards Models , Young Adult
7.
Oncogene ; 33(19): 2454-63, 2014 May 08.
Article in English | MEDLINE | ID: mdl-23752186

ABSTRACT

Despite frequent KRAS mutation, the early molecular mechanisms of pancreatic ductal adenocarcinoma (PDAC) development have not been fully elucidated. By tracking a potential regulator of another feature of PDAC precursors, acquisition of foregut or gastric epithelial gene signature, we herein report that aberrant overexpression of ecotropic viral integration site 1 (EVI1) oncoprotein, which is usually absent in normal pancreatic duct, is a widespread marker across the full spectrum of human PDAC precursors and PDAC. In pancreatic cancer cells, EVI1 depletion caused remarkable inhibition of cell growth and migration, indicating its oncogenic roles. Importantly, we found that EVI1 upregulated KRAS expression through suppression of a potent KRAS suppressor, miR-96, in pancreatic cancer cells. Collectively, the present findings suggest that EVI1 overexpression and KRAS mutation converge on activation of the KRAS pathway in early phases of pancreatic carcinogenesis and propose EVI1 and/or miR-96 as early markers and therapeutic targets in this dismal disease.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , DNA-Binding Proteins/genetics , MicroRNAs/genetics , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogenes/genetics , Transcription Factors/genetics , ras Proteins/genetics , Blotting, Western , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic/genetics , Humans , Immunohistochemistry , MDS1 and EVI1 Complex Locus Protein , Mutation , Oligonucleotide Array Sequence Analysis , Oncogenes , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins p21(ras) , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Transcription Factors/metabolism , Up-Regulation , ras Proteins/metabolism
8.
Oncogene ; 32(33): 3840-5, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-22964630

ABSTRACT

Cancer stem cells are believed to be responsible for tumor initiation and development. Much current research on human brain tumors is focused on the stem-like properties of glioblastoma stem cells (GSCs). However, little is known about the molecular mechanisms of cell cycle regulation that discriminate between GSCs and differentiated glioblastoma cells. Here we show that cyclin D2 is the cyclin that is predominantly expressed in GSCs and suppression of its expression by RNA interference causes G1 arrest in vitro and growth retardation of GSCs xenografted into immunocompromised mice in vivo. We also demonstrate that the expression of cyclin D2 is suppressed upon serum-induced differentiation similar to what was observed for the cancer stem cell marker CD133. Taken together, our results demonstrate that cyclin D2 has a critical role in cell cycle progression and the tumorigenicity of GSCs.


Subject(s)
Cell Cycle/physiology , Cyclin D2/metabolism , Glioblastoma/metabolism , Neoplastic Stem Cells/metabolism , Animals , Cell Line, Tumor , Flow Cytometry , Glioblastoma/pathology , Humans , Immunoblotting , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/pathology , Oligonucleotide Array Sequence Analysis , RNA Interference , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous
9.
Diabetologia ; 55(12): 3318-30, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22983607

ABSTRACT

AIMS/HYPOTHESIS: Insufficient insulin secretion and reduced pancreatic beta cell mass are hallmarks of type 2 diabetes. Here, we focused on a family of serine-threonine kinases known as homeodomain-interacting protein kinases (HIPKs). HIPKs are implicated in the modulation of Wnt signalling, which plays a crucial role in transcriptional activity, and in pancreas development and maintenance. The aim of the present study was to characterise the role of HIPKs in glucose metabolism. METHODS: We used RNA interference to characterise the role of HIPKs in regulating insulin secretion and transcription activity. We conducted RT-PCR and western blot analyses to analyse the expression and abundance of HIPK genes and proteins in the islets of high-fat diet-fed mice. Glucose-induced insulin secretion and beta cell proliferation were measured in islets from Hipk3 ( -/- ) mice, which have impaired glucose tolerance owing to an insulin secretion deficiency. The abundance of pancreatic duodenal homeobox (PDX)-1 and glycogen synthase kinase (GSK)-3ß phosphorylation in Hipk3 ( -/- ) islets was determined by immunohistology and western blot analyses. RESULTS: We found that HIPKs regulate insulin secretion and transcription activity. Hipk3 expression was most significantly increased in the islets of high-fat diet-fed mice. Furthermore, glucose-induced insulin secretion and beta cell proliferation were decreased in the islets of Hipk3 ( -/- ) mice. Levels of PDX1 and GSK-3ß phosphorylation were significantly decreased in Hipk3 ( -/- ) islets. CONCLUSIONS/INTERPRETATION: Depletion of HIPK3 impairs insulin secretion and glucose tolerance. Decreased levels of HIPK3 may play a substantial role in the pathogenesis of type 2 diabetes.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Animals , Cells, Cultured , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/pathology , Diet, High-Fat , Female , Glucose Tolerance Test , Insulin Secretion , Insulin-Secreting Cells/pathology , Male , Mice , Mice, Knockout , Pancreas/metabolism , RNA Interference
10.
Br J Cancer ; 106(1): 126-32, 2012 Jan 03.
Article in English | MEDLINE | ID: mdl-22095227

ABSTRACT

BACKGROUND: Molecular characterisation using gene-expression profiling will undoubtedly improve the prediction of treatment responses, and ultimately, the clinical outcome of cancer patients. METHODS: To establish the procedures to identify responders to FOLFOX therapy, 83 colorectal cancer (CRC) patients including 42 responders and 41 non-responders were divided into training (54 patients) and test (29 patients) sets. Using Random Forests (RF) algorithm in the training set, predictor genes for FOLFOX therapy were identified, which were applied to test samples and sensitivity, specificity, and out-of-bag classification accuracy were calculated. RESULTS: In the training set, 22 of 27 responders (81.4% sensitivity) and 23 of 27 non-responders (85.1% specificity) were correctly classified. To improve the prediction model, we removed the outliers determined by RF, and the model could correctly classify 21 of 23 responders (91.3%) and 22 of 23 non-responders (95.6%) in the training set, and 80.0% sensitivity and 92.8% specificity, with an accuracy of 69.2% in 29 independent test samples. CONCLUSION: Random Forests on gene-expression data for CRC patients was effectively able to stratify responders to FOLFOX therapy with high accuracy, and use of pharmacogenomics in anticancer therapy is the first step in planning personalised therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Models, Theoretical , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Fluorouracil/administration & dosage , Humans , Leucovorin/administration & dosage , Oligonucleotide Array Sequence Analysis , Organoplatinum Compounds/administration & dosage , Survival Analysis
11.
Leukemia ; 25(12): 1849-56, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21738213

ABSTRACT

MicroRNA-125b-1 (miR-125b-1) is a target of a chromosomal translocation t(11;14)(q24;q32) recurrently found in human B-cell precursor acute lymphoblastic leukemia (BCP-ALL). This translocation results in overexpression of miR-125b controlled by immunoglobulin heavy chain gene (IGH) regulatory elements. In addition, we found that six out of twenty-one BCP-ALL patients without t(11;14)(q24;q32) showed overexpression of miR-125b. Interestingly, four out of nine patients with BCR/ABL-positive BCP-ALL and one patient with B-cell lymphoid crisis that had progressed from chronic myelogenous leukemia overexpressed miR-125b. To examine the role of the deregulated expression of miR-125b in the development of B-cell tumor in vivo, we generated transgenic mice mimicking the t(11;14)(q24;q32) (Eµ/miR-125b-TG mice). Eµ/miR-125b-TG mice overexpressed miR-125b driven by IGH enhancer and promoter and developed IgM-negative or IgM-positive lethal B-cell malignancies with clonal proliferation. B cells obtained from the Eµ/miR-125b-TG mice were resistant to apoptosis induced by serum starvation. We identified Trp53inp1, a pro-apoptotic gene induced by cell stress, as a novel target gene of miR-125b in hematopoietic cells in vitro and in vivo. Our results provide direct evidence that miR-125b has important roles in the tumorigenesis of precursor B cells.


Subject(s)
Immunoglobulin mu-Chains/genetics , MicroRNAs/physiology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Animals , Apoptosis , Base Sequence , Blotting, Southern , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 14/genetics , Flow Cytometry , Humans , Luciferases/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , Translocation, Genetic/genetics
12.
Oncogene ; 30(14): 1693-705, 2011 Apr 07.
Article in English | MEDLINE | ID: mdl-21132002

ABSTRACT

Stem cells in normal tissues and cancer-initiating cells (CICs) are known to be enriched in side population (SP) cells. However, the factors responsible for the regulation of expression of ABCG2, involved in efflux of dyes, in SP cells have not been fully investigated. Here, we characterized the SP cells within diffuse-type gastric carcinoma, and examined the effects of transforming growth factor-ß (TGF-ß) on SP cells. Diffuse-type gastric carcinoma cells established from four independent patients universally contained SP cells between 1 and 4% of total cells, which displayed greater tumorigenicity than non-SP cells did. TGF-ß repressed the transcription of ABCG2 through direct binding of Smad2/3 to its promoter/enhancer, and the number of SP cells and the tumor-forming ability of cancer cells were decreased by TGF-ß, although ABCG2 is not directly involved in the tumor-forming ability of SP cells. Cancer cells from metastatic site expressed much higher levels of ABCG2 and included a greater percentage of SP cells than parental cancer cells did. SP cells are thus responsible for the progression of diffuse-type gastric carcinoma, and TGF-ß negatively contributes to maintain the CICs within the cancer.


Subject(s)
Carcinoma/pathology , Neoplastic Stem Cells/drug effects , Side-Population Cells/drug effects , Stomach Neoplasms/pathology , Transforming Growth Factor beta/pharmacology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/biosynthesis , Animals , Cell Line, Tumor , Disease Progression , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lymphatic Metastasis , Mice , Neoplasm Proteins/biosynthesis , Promoter Regions, Genetic/drug effects , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transplantation, Heterologous
13.
Oncogene ; 30(5): 619-30, 2011 Feb 03.
Article in English | MEDLINE | ID: mdl-20890304

ABSTRACT

The androgen receptor (AR) is a critical transcriptional factor that contributes to the development and the progression of prostate cancer (PCa) by regulating the transcription of various target genes. Genome-wide screening of androgen target genes provides useful information to understand a global view of AR-mediated gene network in PCa. In this study, we performed 5'-cap analysis of gene expression (CAGE) to determine androgen-regulated transcription start sites (TSSs) and chromatin immunoprecipitation (ChIP) on array (ChIP-chip) analysis to identify AR binding sites (ARBSs) and histone H3 acetylated (AcH3) sites in the human genome. CAGE determined 13 110 distinct, androgen-regulated TSSs (P<0.01), and ChIP-chip analysis identified 2872 androgen-dependent ARBSs (P<1e-5) and 25 945 AcH3 sites (P<1e-4). Both androgen-regulated coding genes and noncoding RNAs, including microRNAs (miRNAs) were determined as androgen target genes. Besides prototypic androgen-regulated TSSs in annotated gene promoter regions, there are many androgen-dependent TSSs that are widely distributed throughout the genome, including those in antisense (AS) direction of RefSeq genes. Several pairs of sense/antisense promoters were newly identified within single RefSeq gene regions. The integration of CAGE and ChIP-chip analyses successfully identified a cluster of androgen-inducible miRNAs, as exemplified by the miR-125b-2 cluster on chromosome 21. Notably, the number of androgen-upregulated genes was larger in LNCaP cells treated with R1881 for 24 h than for 6 h, and the percentage of androgen-upregulated genes accompanied with adjacent ARBSs was also much higher in cells treated with R1881 for 24 h than 6 h. On the basis of the Oncomine database, the majority of androgen-upregulated genes containing adjacent ARBSs and CAGE tag clusters in our study were previously confirmed as androgen target genes in PCa. The integrated high-throughput genome analyses of CAGE and ChIP-chip provide useful information for elucidating the AR-mediated transcriptional network that contributes to the development and progression of PCa.


Subject(s)
Chromatin Immunoprecipitation/methods , Gene Expression Profiling/methods , Oligonucleotide Array Sequence Analysis/methods , Receptors, Androgen/genetics , Acetylation , Androgens/pharmacology , Binding Sites/genetics , Cell Line, Tumor , Dihydrotestosterone/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Genome, Human/genetics , Genomics/methods , Histones/metabolism , Humans , Male , Promoter Regions, Genetic/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Signal Transduction/genetics , Transcription Initiation Site
14.
Oncogene ; 29(13): 1897-908, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20062086

ABSTRACT

Endometrial cancer is one of the tumor types in which either chromosomal instability (CIN) or microsatellite instability (MSI) may occur. It is known to possess mutations frequently in the Ras-PI3K (phosphatidylinositol 3'-kinase) pathway. We performed a comprehensive genomic survey in 31 endometrial carcinomas with paired DNA for chromosomal imbalances (25 by the 50K and 6 by the 250K single-nucleotide polymorphism (SNP) array), and screened 25 of the 31 samples for MSI status and mutational status in the Ras-PI3K pathway genes. We detected five or more copy number changes (classified as CIN-extensive) in 9 (29%), 1 to 4 changes (CIN-intermediate) in 17 (55%) and no changes (CIN-negative) in 5 (16%) tumors. Positive MSI was less common in CIN-extensive tumors (14%), compared with CIN-intermediate/negative tumors (50%), and multivariate analysis showed that CIN-extensive is an independent poor prognostic factor. SNP array analysis unveiled copy number neutral LOH at 54 loci in 13 tumors (42%), including four at the locus of PTEN. In addition to eight (26%) tumors with PTEN deletions, we detected chromosomal imbalances of NF1, K-Ras and PIK3CA in four (13%), four (13%) and six (19%) tumors, respectively. In all, 7 of the 9 CIN-extensive tumors harbor deletions in the loci of PTEN and/or NF1, whereas all the 10 MSI-positive tumors possess PTEN, PIK3CA and/or K-Ras mutations. Our results showed that genomic alterations in the Ras-PI3K pathway are remarkably widespread in endometrial carcinomas, regardless of the type of genomic instability, and suggest that the degree of CIN is a useful biomarker for prognosis in endometrial carcinomas.


Subject(s)
Chromosomal Instability/genetics , Chromosomes/genetics , Endometrial Neoplasms/genetics , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Polymorphism, Single Nucleotide , Prognosis , Class I Phosphatidylinositol 3-Kinases , Endometrial Neoplasms/diagnosis , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genome, Human , Humans , Mutation , Oligonucleotide Array Sequence Analysis
15.
Br J Cancer ; 102(1): 206-12, 2010 Jan 05.
Article in English | MEDLINE | ID: mdl-19997106

ABSTRACT

BACKGROUND: Tumour-initiating cells (TICs) or cancer stem cells can exist as a small population in malignant tissues. The signalling pathways activated in TICs that contribute to tumourigenesis are not fully understood. METHODS: Several breast cancer cell lines were sorted with CD24 and CD44, known markers for enrichment of breast cancer TICs. Tumourigenesis was analysed using sorted cells and total RNA was subjected to gene expression profiling and gene set enrichment analysis (GSEA). RESULTS: We showed that several breast cancer cell lines have a small population of CD24(-/low)/CD44(+) cells in which TICs may be enriched, and confirmed the properties of TICs in a xenograft model. GSEA revealed that CD24(-/low)/CD44(+) cell populations are enriched for genes involved in transforming growth factor-beta, tumour necrosis factor, and interferon response pathways. Moreover, we found the presence of nuclear factor-kappaB (NF-kappaB) activity in CD24(-/low)/CD44(+) cells, which was previously unrecognised. In addition, NF-kappaB inhibitor dehydroxymethylepoxyquinomicin (DHMEQ) prevented tumourigenesis of CD24(-/low)/CD44(+) cells in vivo. CONCLUSION: Our findings suggest that signalling pathways identified using GSEA help to identify molecular targets and biomarkers for TIC-like cells.


Subject(s)
Breast Neoplasms/pathology , CD24 Antigen/analysis , Cell Separation/methods , Gene Expression Profiling , Hyaluronan Receptors/analysis , Neoplasm Proteins/analysis , Neoplastic Stem Cells/physiology , Animals , Benzamides/pharmacology , Benzamides/therapeutic use , Biomarkers , Breast Neoplasms/genetics , Cyclohexanones/pharmacology , Cyclohexanones/therapeutic use , Female , Genetic Vectors/pharmacology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , NF-kappa B/antagonists & inhibitors , NF-kappa B/physiology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/physiology , Neoplastic Stem Cells/chemistry , Neoplastic Stem Cells/transplantation , Oligonucleotide Array Sequence Analysis , Signal Transduction/physiology , Transduction, Genetic , Xenograft Model Antitumor Assays
16.
Br J Cancer ; 101(1): 145-8, 2009 Jul 07.
Article in English | MEDLINE | ID: mdl-19491896

ABSTRACT

BACKGROUND: The phosphatidylinositol 3'-kinase (PI3K)-AKT pathway is activated in many human cancers and plays a key role in cell proliferation and survival. A mutation (E17K) in the pleckstrin homology domain of the AKT1 results in constitutive AKT1 activation by means of localisation to the plasma membrane. The AKT1 (E17K) mutation has been reported in some tumour types (breast, colorectal, ovarian and lung cancers), and it is of interest which tumour types other than those possess the E17K mutation. METHODS: We analysed the presence of the AKT1 (E17K) mutation in 89 endometrial cancer tissue specimens and in 12 endometrial cancer cell lines by PCR and direct sequencing. RESULTS: We detected two AKT1 (E17K) mutations in the tissue samples (2 out of 89) and no mutations in the cell lines. These two AKT1 mutant tumours do not possess any mutations in PIK3CA, PTEN and K-Ras. INTERPRETATION: Our results and earlier reports suggest that AKT1 mutations might be mutually exclusive with other PI3K-AKT-activating alterations, although PIK3CA mutations frequently coexist with other alterations (such as HER2, K-Ras and PTEN) in several types of tumours.


Subject(s)
Blood Proteins/genetics , Endometrial Neoplasms/genetics , Mutation, Missense , Phosphoproteins/genetics , Proto-Oncogene Proteins c-akt/genetics , Cell Line, Tumor , DNA Methylation , Endometrial Neoplasms/enzymology , Female , Humans , PTEN Phosphohydrolase/biosynthesis , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Polymorphism, Single Nucleotide , Protein Structure, Tertiary , Proto-Oncogene Proteins c-akt/metabolism
17.
Rheumatology (Oxford) ; 47(6): 780-8, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18388148

ABSTRACT

OBJECTIVES: We monitored the mRNA expression profiles of peripheral blood cells during treatment with a TNF-alpha inhibitor, infliximab, in patients with RA. Using a DNA microarray analysis, we demonstrated a unique set of genes, with distinct baseline and post-treatment changes in expression between responders and non-responders to infliximab treatment. METHODS: Using a customized low-density cDNA microarray with 747 genes and a reliable data collection system, we monitored the mRNA expression profiles of whole blood cells from 18 RA patients before and after the infusion of infliximab for up to 22 weeks. The clinical response to treatment with infliximab was determined using the ACR response criteria, the disease activity score of 28 joints (DAS28), and individual clinical parameters. The patients were classified as responders or non-responders based on their ACR50% response at 22 weeks. RESULTS: Approximately 15% of the total genes were found to exhibit a >1.5-fold change, compared with their reference values, at one or more time points during the 22 weeks of infliximab therapy. The expression of inflammatory genes, such as IFN-related genes, was strongly correlated with the serum level of CRP and the DAS28. The increased expression of inflammatory genes in responders was normalized within 2 weeks and then remained at a normal level during the treatment period. In contrast, in the non-responders, the elevated expression at baseline, although it was significantly decreased at 2 weeks, returned to the baseline level after 14 weeks. In addition to inflammatory genes, we identified several groups of genes with distinct differences in expression between the responders and non-responders. CONCLUSIONS: Our results suggest that a customized low-density microarray is useful for monitoring mRNA expression profiles in peripheral blood cells, enabling us to identify a unique set of genes with differentially regulated expressions in responders and non-responders to a TNF inhibitor among patients with RA.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Gene Expression Regulation/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adult , Aged , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/genetics , Biomarkers/blood , C-Reactive Protein/metabolism , Female , Gene Expression Profiling/methods , Humans , Infliximab , Male , Middle Aged , Oligonucleotide Array Sequence Analysis/methods , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Severity of Illness Index , Treatment Outcome
18.
Leukemia ; 21(11): 2246-57, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17690703

ABSTRACT

Transforming growth factor-beta (TGF-beta)-stimulated clone-22 (TSC-22) was originally isolated as a TGF-beta-inducible gene. In this study, we identified TSC-22 as a potential leukemia suppressor. Two types of FMS-like tyrosine kinase-3 (Flt3) mutations are frequently found in acute myeloid leukemia: Flt3-ITD harboring an internal tandem duplication in the juxtamembrane domain associated with poor prognosis and Flt3-TKD harboring a point mutation in the kinase domain. Comparison of gene expression profiles between Flt3-ITD- and Flt3-TKD-transduced Ba/F3 cells revealed that constitutive activation of Flt3 by Flt3-TKD, but not Flt3-ITD, upregulated the expression of TSC-22. Importantly, treatment with an Flt3 inhibitor PKC412 or an Flt3 small interfering RNA decreased the expression level of TSC-22 in Flt3-TKD-transduced cells. Forced expression of TSC-22 suppressed the growth and accelerated the differentiation of several leukemia cell lines into monocytes, in particular, in combination with differentiation-inducing reagents. On the other hand, a dominant-negative form of TSC-22 accelerated the growth of Flt3-TKD-transduced 32Dcl.3 cells. Collectively, these results suggest that TSC-22 is a possible target of leukemia therapy.


Subject(s)
Gene Expression Regulation, Leukemic , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Leukemia/therapy , Repressor Proteins/therapeutic use , fms-Like Tyrosine Kinase 3/chemistry , Animals , HL-60 Cells , Humans , Leukemia/immunology , Male , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , U937 Cells , fms-Like Tyrosine Kinase 3/immunology
19.
Int J Gynecol Cancer ; 17(6): 1300-6, 2007.
Article in English | MEDLINE | ID: mdl-17425679

ABSTRACT

Aldo-keto reductase family 1, member B10 (AKR1B10), an enzyme that converts retinals into retinols is known to detect in non-small cell lung carcinoma (squamous cell- and adeno-carcinomas), but is barely expressed in normal tissues. Since these types of carcinoma occur frequently in the uterus (like in the lung), AKR1B10 may also be overexpressed in two major types of uterine cancer, cervical cancer (CC), and endometrial cancer (EMC). The objective of this study is to investigate AKR1B10 expression in uterine cancer and to analyze its clinical significance. In samples from uterine cancer patients, AKR1B10 was detected in 6 out of 30 (20.0%) CC cases and 6 out of 38 (15.8%) EMC cases. Statistical analysis indicated that AKR1B10 expression was associated with tumor recurrence after surgery and keratinization of squamous cell carcinoma only in CC. Although retinol (a metabolic product by AKR1B10) was observed in the normal epithelium, the molecule was not observed in cancer cells of AKR1B10-positive CC samples suggesting that the recurrence in CC may not depend on the convert of retinals into retinols via AKR1B10, a potential indicator in the management of patients with CC.


Subject(s)
Aldehyde Reductase/metabolism , Biomarkers, Tumor/metabolism , Carcinoma/metabolism , Endometrial Neoplasms/metabolism , Neoplasm Recurrence, Local/metabolism , Uterine Cervical Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Aldo-Keto Reductases , Carcinoma/pathology , Carcinoma/surgery , Case-Control Studies , Cervix Uteri/pathology , Endometrial Neoplasms/pathology , Endometrial Neoplasms/surgery , Endometrium/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Middle Aged , Risk Factors , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/surgery , Vitamin A/metabolism
20.
Oncogene ; 26(30): 4453-63, 2007 Jun 28.
Article in English | MEDLINE | ID: mdl-17297473

ABSTRACT

The androgen receptor (AR) plays a key role as a transcriptional factor in prostate development and carcinogenesis. Identification of androgen-regulated genes is essential to elucidate the AR pathophysiology in prostate cancer. Here, we identified androgen target genes that are directly regulated by AR in LNCaP cells, by combining chromatin immunoprecipitation (ChIP) with tiling microarrays (ChIP-chip). ChIP-enriched or control DNAs from the cells treated with R1881 were hybridized with the ENCODE array, in which a set of regions representing approximately 1% of the whole genome. We chose 10 bona fide AR-binding sites (ARBSs) (P<1e-5) and validated their significant AR recruitment ligand dependently. Eight upregulated genes by R1881 were identified in the vicinity of the ARBSs. Among the upregulated genes, we focused on UGT1A and CDH2 as AR target genes, because the ARBSs close to these genes (in UGT1A distal promoter and CDH2 intron 1) were most significantly associated with acetylated histone H3/H4, RNA polymerase II and p160 family co-activators. Luciferase reporter constructs including those two ARBSs exhibited ligand-dependent transcriptional regulator/enhancer activities. The present study would be powerful to extend our knowledge of the diversity of androgen genetic network and steroid action in prostate cancer cells.


Subject(s)
Androgens/pharmacology , Chromatin Immunoprecipitation/methods , Oligonucleotide Array Sequence Analysis/methods , Prostatic Neoplasms/genetics , Response Elements , Acetylation , Antigens, CD/genetics , Binding Sites , Cadherins/genetics , Cell Line, Tumor , Glucuronosyltransferase/genetics , Histones/metabolism , Humans , Male , RNA Polymerase II/metabolism , Receptors, Androgen/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...