Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Gut ; 69(6): 1039-1052, 2020 06.
Article in English | MEDLINE | ID: mdl-31542774

ABSTRACT

OBJECTIVE: Genomic structural variations (SVs) causing rewiring of cis-regulatory elements remain largely unexplored in gastric cancer (GC). To identify SVs affecting enhancer elements in GC (enhancer-based SVs), we integrated epigenomic enhancer profiles revealed by paired-end H3K27ac ChIP-sequencing from primary GCs with tumour whole-genome sequencing (WGS) data (PeNChIP-seq/WGS). DESIGN: We applied PeNChIP-seq to 11 primary GCs and matched normal tissues combined with WGS profiles of >200 GCs. Epigenome profiles were analysed alongside matched RNA-seq data to identify tumour-associated enhancer-based SVs with altered cancer transcription. Functional validation of candidate enhancer-based SVs was performed using CRISPR/Cas9 genome editing, chromosome conformation capture assays (4C-seq, Capture-C) and Hi-C analysis of primary GCs. RESULTS: PeNChIP-seq/WGS revealed ~150 enhancer-based SVs in GC. The majority (63%) of SVs linked to target gene deregulation were associated with increased tumour expression. Enhancer-based SVs targeting CCNE1, a key driver of therapy resistance, occurred in 8% of patients frequently juxtaposing diverse distal enhancers to CCNE1 proximal regions. CCNE1-rearranged GCs were associated with high CCNE1 expression, disrupted CCNE1 topologically associating domain (TAD) boundaries, and novel TAD interactions in CCNE1-rearranged primary tumours. We also observed IGF2 enhancer-based SVs, previously noted in colorectal cancer, highlighting a common non-coding genetic driver alteration in gastric and colorectal malignancies. CONCLUSION: Integrated paired-end NanoChIP-seq and WGS of gastric tumours reveals tumour-associated regulatory SV in regions associated with both simple and complex genomic rearrangements. Genomic rearrangements may thus exploit enhancer-hijacking as a common mechanism to drive oncogene expression in GC.


Subject(s)
Adenocarcinoma/metabolism , Cyclin E/metabolism , Enhancer Elements, Genetic/genetics , Insulin-Like Growth Factor II/metabolism , Oncogene Proteins/metabolism , Stomach Neoplasms/metabolism , Adenocarcinoma/genetics , Genomic Structural Variation/genetics , Humans , Stomach Neoplasms/genetics , Whole Genome Sequencing
2.
Gastroenterology ; 153(1): 191-204.e16, 2017 07.
Article in English | MEDLINE | ID: mdl-28390866

ABSTRACT

BACKGROUND & AIMS: Fibroblasts that interact with cancer cells are called cancer-associated fibroblasts (CAFs), which promote progression of different tumor types. We investigated the characteristics and functions of CAFs in diffuse-type gastric cancers (DGCs) by analyzing features of their genome and gene expression patterns. METHODS: We isolated CAFs and adjacent non-cancer fibroblasts (NFs) from 110 gastric cancer (GC) tissues from patients who underwent gastrectomy in Japan from 2008 through 2016. Cells were identified using specific markers of various cell types by immunoblot and flow cytometry. We selected pairs of CAFs and NFs for whole-exome and RNA sequencing analyses, and compared expression of specific genes using quantitative reverse transcription PCR. Protein levels and phosphorylation were compared by immunoblot and immunofluorescence analyses. Rhomboid 5 homolog 2 (RHBDF2) was overexpressed from a transgene in fibroblasts or knocked down using small interfering RNAs. Motility and invasiveness of isolated fibroblasts and GC cell lines (AGS, KATOIII, MKN45, NUGC3, NUGC4, OCUM-2MD3 and OCUM-12 cell lines) were quantified by real-time imaging analyses. We analyzed 7 independent sets of DNA microarray data from patients with GC and associated expression levels of specific genes with patient survival times. Nude mice were given injections of OCUM-2MD3 in the stomach wall; tumors and metastases were collected and analyzed by immunohistochemistry. RESULTS: Many of the genes with increased expression in CAFs compared with NFs were associated with transforming growth factor beta 1 (TGFB1) activity. When CAFs were cultured in extracellular matrix, they became more motile than NFs; DGC cells incubated with CAFs were also more motile and invasive in vitro than DGC cells not incubated with CAFs. When injected into nude mice, CAF-incubated DGC cells invaded a greater number of lymphatic vessels than NF-incubated DGC cells. We identified RHBDF2 as a gene overexpressed in CAFs compared with NFs. Knockdown of RHBDF2 in CAFs reduced their elongation and motility in response to TGFB1, whereas overexpression of RHBDF2 in NFs increased their motility in extracellular matrix. RHBDF2 appeared to regulate oncogenic and non-canonical TGFB1 signaling. Knockdown of RHBDF2 in CAFs reduced cleavage of the TGFB receptor 1 (TGFBR1) by ADAM metallopeptidase domain 17 (ADAM17 or TACE) and reduced expression of genes that regulate motility. Incubation of NFs with in interleukin 1 alpha (IL1A), IL1B or tumor necrosis factor, secreted by DGCs, increased fibroblast expression of RHBDF2. Simultaneous high expression of these cytokines in GC samples was associated with shorter survival times of patients. CONCLUSIONS: In CAFs isolated from human DGCs, we observed increased expression of RHBDF2, which regulates TGFB1 signaling. Expression of RHBDF2 in fibroblasts is induced by inflammatory cytokines (such as IL1A, IL1B, and tumor necrosis factor) secreted by DGCs. RHBDF2 promotes cleavage of TGFBR1 by activating TACE and motility of CAFs in response to TGFB1. These highly motile CAFs induce DGCs to invade extracellular matrix and lymphatic vessels in nude mice.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Stomach Neoplasms/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , ADAM17 Protein/metabolism , Animals , Carrier Proteins/analysis , Carrier Proteins/antagonists & inhibitors , Cell Line, Tumor , Cell Movement/genetics , Coculture Techniques , Exome , Extracellular Matrix , Female , Gene Expression , Humans , Interleukin-1alpha/pharmacology , Interleukin-1beta/pharmacology , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Transplantation , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Sequence Analysis, RNA , Signal Transduction/genetics , Stomach Neoplasms/chemistry , Stomach Neoplasms/pathology , Survival Rate , Transcriptome , Tumor Necrosis Factor-alpha/pharmacology
3.
JCI Insight ; 1(9): e87607, 2016 06 16.
Article in English | MEDLINE | ID: mdl-27699276

ABSTRACT

Novel, tumor-specific drugs are urgently needed for a breakthrough in cancer therapy. Herein, we generated a first-in-class humanized antibody (PRL3-zumab) against PRL-3, an intracellular tumor-associated phosphatase upregulated in multiple human cancers, for unconventional cancer immunotherapies. We focused on gastric cancer (GC), wherein elevated PRL-3 mRNA levels significantly correlated with shortened overall survival of GC patients. PRL-3 protein was overexpressed in 85% of fresh-frozen clinical gastric tumor samples examined but not in patient-matched normal gastric tissues. Using human GC cell lines, we demonstrated that PRL3-zumab specifically blocked PRL-3+, but not PRL-3-, orthotopic gastric tumors. In this setting, PRL3-zumab had better therapeutic efficacy as a monotherapy, rather than simultaneous combination with 5-fluorouracil or 5-fluorouracil alone. PRL3-zumab could also prevent PRL-3+ tumor recurrence. Mechanistically, we found that intracellular PRL-3 antigens could be externalized to become "extracellular oncotargets" that serve as bait for PRL3-zumab binding to potentially bridge and recruit immunocytes into tumor microenvironments for killing effects on cancer cells. In summary, our results document a comprehensive cancer therapeutic approach to specific antibody-targeted therapy against the PRL-3 oncotarget as a case study for developing antibodies against other intracellular targets in drug discovery.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Neoplasm Proteins/immunology , Protein Tyrosine Phosphatases/immunology , Stomach Neoplasms/therapy , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Inbred BALB C , Neoplasm Recurrence, Local , Xenograft Model Antitumor Assays
4.
Nat Commun ; 7: 12983, 2016 Sep 28.
Article in English | MEDLINE | ID: mdl-27677335

ABSTRACT

Regulatory enhancer elements in solid tumours remain poorly characterized. Here we apply micro-scale chromatin profiling to survey the distal enhancer landscape of primary gastric adenocarcinoma (GC), a leading cause of global cancer mortality. Integrating 110 epigenomic profiles from primary GCs, normal gastric tissues and cell lines, we highlight 36,973 predicted enhancers and 3,759 predicted super-enhancers respectively. Cell-line-defined super-enhancers can be subclassified by their somatic alteration status into somatic gain, loss and unaltered categories, each displaying distinct epigenetic, transcriptional and pathway enrichments. Somatic gain super-enhancers are associated with complex chromatin interaction profiles, expression patterns correlated with patient outcome and dense co-occupancy of the transcription factors CDX2 and HNF4α. Somatic super-enhancers are also enriched in genetic risk SNPs associated with cancer predisposition. Our results reveal a genome-wide reprogramming of the GC enhancer and super-enhancer landscape during tumorigenesis, contributing to dysregulated local and regional cancer gene expression.

5.
Oncotarget ; 7(6): 6693-710, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26760764

ABSTRACT

Despite the approval of numerous molecular targeted drugs, long-term antiproliferative efficacy is rarely achieved and therapy resistance remains a central obstacle of cancer care. Combined inhibition of multiple cancer-driving pathways promises to improve antiproliferative efficacy. HIF-1 is a driver of gastric cancer and considered to be an attractive target for therapy. We noted that gastric cancer cells are able to functionally compensate the stable loss of HIF-1α. Via transcriptomics we identified a group of upregulated genes in HIF-1α-deficient cells and hypothesized that these genes confer survival upon HIF-1α loss. Strikingly, simultaneous knock-down of HIF-1α and Annexin A1 (ANXA1), one of the identified genes, resulted in complete cessation of proliferation. Using stable isotope-resolved metabolomics, oxidative and reductive glutamine metabolism was found to be significantly impaired in HIF-1α/ANXA1-deficient cells, potentially explaining the proliferation defect. In summary, we present a conceptually novel application of stable gene inactivation enabling in-depth deconstruction of resistance mechanisms. In theory, this experimental approach is applicable to any cancer-driving gene or pathway and promises to identify various new targets for combination therapies.


Subject(s)
Annexin A1/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Stomach Neoplasms/metabolism , Animals , Annexin A1/genetics , Cell Line, Tumor , Cell Proliferation/physiology , Female , Heterografts , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunohistochemistry , Mice , Mice, Inbred NOD , Mice, SCID , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics
6.
Cell Oncol (Dordr) ; 39(1): 35-45, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26650484

ABSTRACT

BACKGROUND: The presence of cancer stem cells (CSCs) is currently regarded as one of the main culprits of tumor formation and therapy failure. It is known that chronic inflammation is associated with CSCs, but it is not clear yet how inflammation affects the development of CSCs. In the present study we aimed to examine the relationship between cancer cell stimulation mediated by immune cells and the acquisition of a CSC-like phenotype. METHODS: Cancer cells derived from single hepatocarcinoma HepG2 cells were treated with mouse splenic B cells (MSBCs) and mouse peritoneal macrophage cells (MPMCs), respectively. The stem cell-like characteristics of the resulting HepG2 cells (MSBC-HepG2 and MPMC-HepG2) were evaluated using different assays, including biomarker assays, in vitro tumoroid and colony forming assays, in vivo tumor forming assays and signal transduction pathway activation assays. RESULTS: Various stemness characteristics of HepG2 cells, including self-renewal, proliferation, chemoresistance and tumorigenicity were evaluated. The expression levels of stemness-related genes and its encoded proteins in the MSBC-HepG2 and MPMC-HepG2 cells were assessed using RT-PCR and FACS analyses. We found that MSBC-HepG2 and MPMC-HepG2 cells possess hepatic CSC properties, including persistent self-renewal, extensive proliferation, drug resistance, high tumorigenic capacity and over-expression of CSC-related genes and proteins (i.e., EpCAM, ALDH, CD133 and CD44), compared to the parental cells. We also found that 1x10(3) MSBC-HepG2 and MPMC-HepG2 cells were able to form tumors in NOD/SCID mice and that the Notch and SHH signaling pathways were highly activated in MSBC-HepG2 cells. CONCLUSIONS: We conclude that the immune system may have a double-edge effect on cancer development. On one hand, immune cells such as B lymphocytes and macrophages may recognize, attack and eliminate cancer cells, whereas on the other hand, they may promote a subset of cancer cells to acquire stem cell-like characteristics.


Subject(s)
B-Lymphocytes/cytology , Macrophages, Peritoneal/cytology , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/pathology , Animals , Apoptosis/drug effects , B-Lymphocytes/drug effects , Cell Proliferation/drug effects , Cell Self Renewal/drug effects , Culture Media, Serum-Free/pharmacology , Female , Hedgehog Proteins/metabolism , Hep G2 Cells , Humans , Macrophages, Peritoneal/drug effects , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/drug effects , Receptors, Notch/metabolism , Signal Transduction/drug effects , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Spleen/cytology
7.
Carcinogenesis ; 36(4): 441-51, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25742747

ABSTRACT

Gastric cancer (GC) is the second leading cause of global cancer mortality worldwide. However, the molecular mechanism underlying its carcinogenesis and drug resistance is not well understood. To identify novel functionally important genes that were differentially expressed due to combinations of genetic and epigenetic changes, we analyzed datasets containing genome-wide mRNA expression, DNA copy number alterations and DNA methylation status from 154 primary GC samples and 47 matched non-neoplastic mucosa tissues from Asian patients. We used concepts of 'within' and 'between' statistical analysis to compare the difference between tumors and controls within each platform, and assessed the correlations between platforms. This 'multi-regulated gene (MRG)' analysis identified 126 differentially expressed genes that underwent a combination of copy number and DNA methylation changes. Most genes were located at genomic loci associated with GC. Statistical enrichment analysis showed that MRGs were enriched for cancer, GC and drug response. We analysed several MRGs that previously had not been associated with GC. Knockdown of DDX27, TH1L or IDH3G sensitized cells to epirubicin or cisplatin, and knockdown of RAI14 reduced cell proliferation. Further studies showed that overexpression of DDX27 reduced epirubicin-induced DNA damage and apoptosis. Levels of DDX27 mRNA and protein were increased in early-stage gastric tumors, and may be a potential diagnostic and prognostic marker for GC. In summary, we used an integrative bioinformatics strategy to identify novel genes that are altered in GC and regulate resistance of GC cells to drugs in vitro.


Subject(s)
Antineoplastic Agents/pharmacology , DEAD-box RNA Helicases/genetics , Drug Resistance, Neoplasm/genetics , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Apoptosis/drug effects , Apoptosis/genetics , Calcium-Binding Proteins , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cisplatin/pharmacology , Cytoskeletal Proteins/genetics , DEAD-box RNA Helicases/biosynthesis , DNA Copy Number Variations/genetics , DNA Damage/drug effects , DNA Damage/genetics , DNA Methylation/genetics , Databases, Nucleic Acid , Epirubicin/pharmacology , Gastric Mucosa/cytology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Histones/genetics , Humans , Nerve Tissue Proteins/genetics , Prognosis , RNA Interference , RNA, Small Interfering , Retrospective Studies , Transcription Factors/genetics
8.
Gut ; 64(11): 1721-31, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25385008

ABSTRACT

OBJECTIVE: Differences in gastric cancer (GC) clinical outcomes between patients in Asian and non-Asian countries has been historically attributed to variability in clinical management. However, recent international Phase III trials suggest that even with standardised treatments, GC outcomes differ by geography. Here, we investigated gene expression differences between Asian and non-Asian GCs, and if these molecular differences might influence clinical outcome. DESIGN: We compared gene expression profiles of 1016 GCs from six Asian and three non-Asian GC cohorts, using a two-stage meta-analysis design and a novel biostatistical method (RUV-4) to adjust for technical variation between cohorts. We further validated our findings by computerised immunohistochemical analysis on two independent tissue microarray (TMA) cohorts from Asian and non-Asian localities (n=665). RESULTS: Gene signatures differentially expressed between Asians and non-Asian GCs were related to immune function and inflammation. Non-Asian GCs were significantly enriched in signatures related to T-cell biology, including CTLA-4 signalling. Similarly, in the TMA cohorts, non-Asian GCs showed significantly higher expression of T-cell markers (CD3, CD45R0, CD8) and lower expression of the immunosuppressive T-regulatory cell marker FOXP3 compared to Asian GCs (p<0.05). Inflammatory cell markers CD66b and CD68 also exhibited significant cohort differences (p<0.05). Exploratory analyses revealed a significant relationship between tumour immunity factors, geographic locality-specific prognosis, and postchemotherapy outcomes. CONCLUSIONS: Analyses of >1600 GCs suggest that Asian and non-Asian GCs exhibit distinct tumour immunity signatures related to T-cell function. These differences may influence geographical differences in clinical outcome, and the design of future trials particularly in immuno-oncology.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/immunology , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , Transcriptome , Adenocarcinoma/drug therapy , Adult , Aged , Aged, 80 and over , Asian People/genetics , Female , Humans , Male , Middle Aged , Retrospective Studies , Stomach Neoplasms/drug therapy , Treatment Outcome , Young Adult
9.
Nat Commun ; 5: 4378, 2014 Jul 09.
Article in English | MEDLINE | ID: mdl-25007141

ABSTRACT

Opisthorchiasis is a neglected, tropical disease caused by the carcinogenic Asian liver fluke, Opisthorchis viverrini. This hepatobiliary disease is linked to malignant cancer (cholangiocarcinoma, CCA) and affects millions of people in Asia. No vaccine is available, and only one drug (praziquantel) is used against the parasite. Little is known about O. viverrini biology and the diseases that it causes. Here we characterize the draft genome (634.5 Mb) and transcriptomes of O. viverrini, elucidate how this fluke survives in the hostile environment within the bile duct and show that metabolic pathways in the parasite are highly adapted to a lipid-rich diet from bile and/or cholangiocytes. We also provide additional evidence that O. viverrini and other flukes secrete proteins that directly modulate host cell proliferation. Our molecular resources now underpin profound explorations of opisthorchiasis/CCA and the design of new interventions.


Subject(s)
Bile Ducts, Intrahepatic/parasitology , DNA, Helminth/genetics , Genome/genetics , Opisthorchis/genetics , Opisthorchis/physiology , Amino Acid Sequence , Animals , Bile Duct Neoplasms/parasitology , Bile Duct Neoplasms/physiopathology , Cholangiocarcinoma/parasitology , Cholangiocarcinoma/physiopathology , Helminth Proteins/genetics , Helminth Proteins/physiology , Host-Parasite Interactions/genetics , Host-Parasite Interactions/physiology , Humans , Molecular Sequence Data , Opisthorchiasis/complications , Opisthorchiasis/genetics , Opisthorchiasis/physiopathology
10.
Nat Commun ; 5: 4361, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-25008978

ABSTRACT

Chromatin alterations are fundamental hallmarks of cancer. To study chromatin alterations in primary gastric adenocarcinomas, we perform nanoscale chromatin immunoprecipitation sequencing of multiple histone modifications in five gastric cancers and matched normal tissues. We identify hundreds of somatically altered promoters and predicted enhancers. Many cancer-associated promoters localize to genomic sites lacking previously annotated transcription start sites (cryptic promoters), driving expression of nearby genes involved in gastrointestinal cancer, embryonic development and tissue specification. Cancer-associated promoters overlap with embryonic stem cell regions targeted by polycomb repressive complex 2, exhibiting promoter bivalency and DNA methylation loss. We identify somatically acquired elements exhibiting germline allelic biases and non-coding somatic mutations creating new promoters. Our findings demonstrate the feasibility of profiling chromatin from solid tumours with limited tissue to identify regulatory elements, transcriptional patterns and regulatory genetic variants associated with cancer.


Subject(s)
Adenocarcinoma/genetics , Chromatin/genetics , DNA Fingerprinting/methods , Nanotechnology/methods , Promoter Regions, Genetic/genetics , Regulatory Elements, Transcriptional/genetics , Stomach Neoplasms/genetics , Adenocarcinoma/pathology , Alleles , Case-Control Studies , DNA/genetics , DNA Methylation , DNA, Neoplasm/genetics , Histones/genetics , Humans , Mutation/genetics , Stomach Neoplasms/pathology , Transcription Initiation Site
11.
Breast Cancer Res Treat ; 128(2): 505-16, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21258862

ABSTRACT

Mammographic density (MD) adjusted for age and body mass index (BMI) is a strong heritable breast cancer risk factor; however, its biological basis remains elusive. Previous studies assessed MD-associated histology using random sampling approaches, despite evidence that high and low MD areas exist within a breast and are negatively correlated with respect to one another. We have used an image-guided approach to sample high and low MD tissues from within individual breasts to examine the relationship between histology and degree of MD. Image-guided sampling was performed using two different methodologies on mastectomy tissues (n = 12): (1) sampling of high and low MD regions within a slice guided by bright (high MD) and dark (low MD) areas in a slice X-ray film; (2) sampling of high and low MD regions within a whole breast using a stereotactically guided vacuum-assisted core biopsy technique. Pairwise analysis accounting for potential confounders (i.e. age, BMI, menopausal status, etc.) provides appropriate power for analysis despite the small sample size. High MD tissues had higher stromal (P = 0.002) and lower fat (P = 0.002) compositions, but no evidence of difference in glandular areas (P = 0.084) compared to low MD tissues from the same breast. High MD regions had higher relative gland counts (P = 0.023), and a preponderance of Type I lobules in high MD compared to low MD regions was observed in 58% of subjects (n = 7), but did not achieve significance. These findings clarify the histologic nature of high MD tissue and support hypotheses regarding the biophysical impact of dense connective tissue on mammary malignancy. They also provide important terms of reference for ongoing analyses of the underlying genetics of MD.


Subject(s)
Breast Neoplasms/diagnostic imaging , Breast Neoplasms/prevention & control , Breast/anatomy & histology , Breast/pathology , Mammography , Stromal Cells/pathology , Adult , Cohort Studies , Female , Follow-Up Studies , Humans , Middle Aged , Prognosis , Prospective Studies , Stromal Cells/diagnostic imaging , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL
...