Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Oncotarget ; 7(23): 34977-87, 2016 Jun 07.
Article in English | MEDLINE | ID: mdl-27144336

ABSTRACT

The methylation status of a promoter influences gene expression and aberrant methylation during tumor development has important functional consequences for pancreatic and other cancers. Using methylated CpG island amplification and promoter microarrays, we identified ANK1 as hypomethylated in pancreatic cancers. Expression analysis determined ANK1 as commonly overexpressed in pancreatic cancers relative to normal pancreas. ANK1 was co-expressed with miR-486 in pancreatic cancer cells. Stable knockdown of ANK1 in the pancreatic cancer cell line AsPC1 led to changes in cell morphology, and decreases in colony formation. Stable knockdown of ANK1 also marked reduced the growth of tumors in athymic nude mice. Among patients undergoing pancreaticoduodenectomy, those with pancreatic cancers expressing ANK1 had a poorer prognosis than those without ANK1 expression. These findings indicate a role for ANK1 overexpression in mediating pancreatic cancer tumorigenicity.


Subject(s)
Ankyrins/metabolism , Carcinoma, Pancreatic Ductal/pathology , Gene Expression Regulation, Neoplastic/physiology , Pancreatic Neoplasms/pathology , Adult , Aged , Animals , Biomarkers, Tumor/analysis , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/mortality , DNA Methylation , Female , Heterografts , Humans , Kaplan-Meier Estimate , Male , Mice , Mice, Nude , MicroRNAs , Middle Aged , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/mortality
2.
Gan To Kagaku Ryoho ; 41(6): 773-5, 2014 Jun.
Article in Japanese | MEDLINE | ID: mdl-25129093

ABSTRACT

A 50-year-old female patient underwent distal gastrectomy and intraperitoneal CDDP administration for advanced gastric cancer accompanied by severe peritoneal dissemination. She valued her quality of life and chose an oral anticancer drug, S-1, as a postoperative chemotherapy agent. S-1 was administered at a dose of 100mg/body/day for 4 weeks, followed by a 2- week rest. There were no adverse events due to S-1 and no exacerbation of peritoneal dissemination in the 5 years following surgery. The S-1 administration schedule was then changed to alternate-day administration. Eight years after the surgery, the patient discontinued S-1 treatment and has since survived for 11 years with no obvious cancer recurrence.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Oxonic Acid/therapeutic use , Peritoneal Neoplasms/drug therapy , Stomach Neoplasms/drug therapy , Tegafur/therapeutic use , Combined Modality Therapy , Drug Combinations , Female , Gastrectomy , Humans , Lymph Node Excision , Lymphatic Metastasis , Middle Aged , Neoplasm Invasiveness , Peritoneal Neoplasms/secondary , Stomach Neoplasms/pathology , Stomach Neoplasms/surgery , Treatment Outcome
3.
Oncotarget ; 5(9): 2575-87, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24810906

ABSTRACT

To identify potentially important genes dysregulated in pancreatic cancer, we analyzed genome-wide transcriptional analysis of pancreatic cancers and normal pancreatic duct samples and identified the transcriptional coactivator, EYA2 (Drosophila Eyes Absent Homologue-2) as silenced in the majority of pancreatic cancers. We investigated the role of epigenetic mechanisms of EYA2 gene silencing in pancreatic cancers, performed in vitro and in vivo proliferation and migration assays to assess the effect of EYA2 silencing on tumor cell growth and metastasis formation, and expression analysis to identify genes transcriptionally regulated by EYA2. We found loss of tumoral Eya2 expression in 63% of pancreatic cancers (120/189 cases). Silencing of EYA2 expression in pancreatic cancer cell lines correlated with promoter methylation and histone deacetylation and was reversible with DNA methyltransferase and HDAC inhibitors. EYA2 knockdown in pancreatic cancer cell lines increased cell proliferation. Compared to parental pancreatic cancer cells, pancreatic cancers stably-expressing EYA2 grew more slowly and had fewer metastases in orthotopic models. The transcriptional changes after stable expression of EYA2 in pancreatic cancer cells included induction of genes in the TGFbeta pathway. Epigenetic silencing of EYA2 is a common event in pancreatic cancers and stable expression EYA2 limits the growth and metastases of pancreatic adenocarcinoma.


Subject(s)
Adenocarcinoma/secondary , Cell Proliferation , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Gene Silencing , Intracellular Signaling Peptides and Proteins/genetics , Nuclear Proteins/genetics , Pancreatic Neoplasms/pathology , Protein Tyrosine Phosphatases/genetics , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Animals , Base Sequence , Blotting, Western , Cell Cycle , Cell Movement , Chromatin Immunoprecipitation , DNA Methylation , Flow Cytometry , Humans , Immunoenzyme Techniques , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Nude , Molecular Sequence Data , Neoplasm Metastasis , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Pancreas/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Promoter Regions, Genetic/genetics , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein Tyrosine Phosphatases/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
World J Surg Oncol ; 10: 215, 2012 Oct 10.
Article in English | MEDLINE | ID: mdl-23046863

ABSTRACT

We report a rare case in which hemothorax occurred in addition to hemoperitoneum due to spontaneous rupture of hepatocellular carcinoma (HCC) originating from the caudate lobe of the liver. The case pertains to a 56-year-old female who was transported to our hospital for impaired consciousness due to hemorrhagic shock. Computed tomography (CT) demonstrated ruptured HCC originating from the caudate lobe and accompanying hemoperitoneum and right hemothorax. Hemostasis was carried out by transcatheter arterial embolization (TAE), and surgery was conducted approximately one month after TAE. In the present case, no lesions as possible sources of bleeding were observed inside the pleural cavity, and, moreover, the diaphragm had no abnormalities in the intraoperative findings, suggesting that blood from the ruptured tumor may have traversed the intact diaphragm to enter the right pleural cavity soon after the HCC rupture. However, to the best of our knowledge, no similar cases of HCC have been reported to date, and this case is thus believed to be very rare. This unusual phenomenon may therefore be strongly associated with the location of the ruptured tumor and the formation of a hematoma inside the omental bursa. We discuss the mechanism causing hemothorax in the present case and also review the previously reported cases of ruptured HCC complicated by hemothorax.


Subject(s)
Carcinoma, Hepatocellular/complications , Hemoperitoneum/pathology , Hemothorax/etiology , Liver Neoplasms/complications , Rupture, Spontaneous/etiology , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/surgery , Female , Hemoperitoneum/surgery , Hemothorax/pathology , Hemothorax/surgery , Humans , Liver Neoplasms/pathology , Liver Neoplasms/surgery , Middle Aged , Review Literature as Topic , Rupture, Spontaneous/pathology , Rupture, Spontaneous/surgery , Tomography, X-Ray Computed
5.
PLoS One ; 7(9): e43456, 2012.
Article in English | MEDLINE | ID: mdl-22984426

ABSTRACT

PURPOSE: Cancer associated stromal fibroblasts (CAFs) undergo transcriptional and phenotypic changes that contribute to tumor progression, but the mechanisms responsible for these changes are not well understood. Aberrant DNA methylation is an important cause of transcriptional alterations in cancer cells but it is not known how important DNA methylation alterations are to CAF behavior. EXPERIMENTAL DESIGN: We used Affymetrix exon arrays to compare genes induced by the DNA methylation inhibitor 5-aza-dC in cultured pancreatic cancer associated fibroblasts, pancreatic control fibroblasts and pancreatic cancer cell lines. RESULTS: We found that pancreatic CAFs and control pancreatic fibroblasts were less responsive to 5-aza-dC-mediated gene reactivation than pancreatic cancer cells (mean+/-SD of genes induced ≥ 5-fold was 9 ± 10 genes in 10 pancreatic CAF cultures, 17 ± 14 genes in 3 control pancreatic fibroblast cultures, and 134 ± 85 genes in 4 pancreatic cancer cell lines). We examined differentially expressed genes between CAFs and control fibroblasts for candidate methylated genes and identified the disintegrin and metalloprotease, ADAM12 as hypomethylated and overexpressed in pancreatic CAF lines and overexpressed in fibroblasts adjacent to primary pancreatic adenocarcinomas. CONCLUSIONS: Compared to pancreatic cancer cells, few genes are reactivated by DNMT1 inhibition in pancreatic CAFs suggesting these cells do not harbor many functionally important alterations in DNA methylation. CAFs may also not be very responsive to therapeutic targeting with DNA methylation inhibitors.


Subject(s)
Azacitidine/pharmacology , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation, Neoplastic/drug effects , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAM12 Protein , Base Sequence , Blotting, Western , Cell Line, Tumor , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , Female , Fibroblasts/drug effects , Genes, Neoplasm/genetics , Genetic Association Studies , Humans , Immunohistochemistry , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Middle Aged , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Analysis, DNA , Up-Regulation/drug effects , Up-Regulation/genetics
6.
Mol Cancer Ther ; 11(9): 1999-2009, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22752426

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy, with most patients facing an adverse clinical outcome. Aberrant Notch pathway activation has been implicated in the initiation and progression of PDAC, specifically the aggressive phenotype of the disease. We used a panel of human PDAC cell lines as well as patient-derived PDAC xenografts to determine whether pharmacologic targeting of Notch pathway could inhibit PDAC growth and potentiate gemcitabine sensitivity. MRK-003, a potent and selective γ-secretase inhibitor, treatment resulted in the downregulation of nuclear Notch1 intracellular domain, inhibition of anchorage-independent growth, and reduction of tumor-initiating cells capable of extensive self-renewal. Pretreatment of PDAC cells with MRK-003 in cell culture significantly inhibited the subsequent engraftment in immunocompromised mice. MRK-003 monotherapy significantly blocked tumor growth in 5 of 9 (56%) PDAC xenografts. A combination of MRK-003 and gemcitabine showed enhanced antitumor effects compared with gemcitabine in 4 of 9 (44%) PDAC xenografts, reduced tumor cell proliferation, and induced both apoptosis and intratumoral necrosis. Gene expression analysis of untreated tumors indicated that upregulation of NF-κB pathway components was predictive of sensitivity to MRK-003, whereas upregulation in B-cell receptor signaling and nuclear factor erythroid-derived 2-like 2 pathway correlated with response to the combination of MRK-003 with gemcitabine. Our findings strengthen the rationale for small-molecule inhibition of Notch signaling as a therapeutic strategy in PDAC.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Cyclic S-Oxides/pharmacology , Pancreatic Neoplasms/drug therapy , Thiadiazoles/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Adhesion , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cyclic S-Oxides/therapeutic use , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Drug Synergism , Gene Expression/drug effects , Humans , Male , Mice , Mice, Nude , Neoplastic Stem Cells/drug effects , Oligonucleotide Array Sequence Analysis , Pancreatic Neoplasms/pathology , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Thiadiazoles/therapeutic use , Transcriptome/drug effects , Xenograft Model Antitumor Assays , Gemcitabine
7.
J Exp Clin Cancer Res ; 31: 55, 2012 Jun 06.
Article in English | MEDLINE | ID: mdl-22672897

ABSTRACT

PURPOSE: It is still technically difficult to collect high purity cancer cells from tumor tissues, which contain noncancerous cells. We hypothesized that xenograft models of NOG mice expressing enhanced green fluorescent protein (eGFP), referred to as NOG-EGFP mice, may be useful for obtaining such high purity cancer cells for detailed molecular and cellular analyses. METHODS: Pancreato-biliary cancer cell lines were implanted subcutaneously to compare the tumorigenicity between NOG-EGFP mice and nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice. To obtain high purity cancer cells, the subcutaneous tumors were harvested from the mice and enzymatically dissociated into single-cell suspensions. Then, the cells were sorted by fluorescence-activated cell sorting (FACS) for separation of the host cells and the cancer cells. Thereafter, the contamination rate of host cells in collected cancer cells was quantified by using FACS analysis. The viability of cancer cells after FACS sorting was evaluated by cell culture and subsequent subcutaneous reimplantation in NOG-EGFP mice. RESULTS: The tumorigenicity of NOG-EGFP mice was significantly better than that of NOD/SCID mice in all of the analyzed cell lines (p < 0.01). Sorting procedures enabled an almost pure collection of cancer cells with only slight contamination by host cells. Reimplantation of the sorted cancer cells formed tumors again, which demonstrated that cell viability after sorting was well maintained. CONCLUSIONS: This method provides a novel cancer sampling system for molecular and cellular analysis with high accuracy and should contribute to the development of personalized medicine.


Subject(s)
Bile Duct Neoplasms/metabolism , Cholangiocarcinoma/metabolism , Green Fluorescent Proteins/genetics , Pancreatic Neoplasms/metabolism , Animals , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic , Cell Line , Cell Survival , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , Disease Models, Animal , Female , Flow Cytometry , Green Fluorescent Proteins/metabolism , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Phenotype
8.
Clin Cancer Res ; 18(16): 4303-12, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22723370

ABSTRACT

PURPOSE: Characterizing the earliest chromosomal alterations of pancreatic precursor neoplasms from individuals with a familial aggregation of pancreatic cancer may provide clues as to the loci of pancreatic cancer susceptibility genes. EXPERIMENTAL DESIGN: We used Illumina 370/660K SNP arrays to conduct genome-wide copy number analysis in 60 benign neoplasms [58 mostly low-grade pancreatic intraepithelial neoplasias (PanIN) and intraductal papillary mucinous neoplasms (IPMN) and two pancreatic neuroendocrine tumors (PNET)] and matched normal tissues from 16 individuals with a family history of pancreatic cancer. PanINs and IPMNs were analyzed for KRAS codon 12/13 mutations. RESULTS: Of 40 benign neoplasms with adequate SNP calls and allele ratios, somatic chromosomal copy number changes were identifiable in only nine lesions, including eight of the 38 PanIN/IPMNs (two of which had identical alterations) and one of the two PNETs. Only two precursor lesions had more than one somatic copy number alteration. In contrast, the overwhelming majority (∼95%) of PanINs harbored KRAS mutations. The chromosomal alterations identified included nine chromosomal arms affected by chromosomal loss and two by chromosomal gain. Copy number loss spanning 9p21.3 was identified in three precursor lesions; two precursors had chromosomal losses affecting 6q and 17p. CONCLUSIONS: Low- and intermediate-grade PanINs and IPMNs from patients with a family history of pancreatic cancer harbor few if any somatic chromosomal alterations. The absence of a locus of recurrent chromosomal loss in most low-grade pancreatic cancer precursor lesions supports the hypothesis that there is no one tumor suppressor gene locus consistently involved in initiating familial pancreatic neoplasia.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , DNA Copy Number Variations , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Aged , Family , Female , Genes, ras , Genome-Wide Association Study , Humans , Loss of Heterozygosity , Male , Middle Aged , Mutation , Neoplasm Grading
9.
Clin Cancer Res ; 18(3): 700-12, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22173550

ABSTRACT

PURPOSE: Intraductal papillary mucinous neoplasms (IPMN) are precursors to infiltrating pancreatic ductal adenocarcinomas. Widespread epigenetic alterations are characteristic of many cancers, yet few studies have systematically analyzed epigenetic alterations of neoplastic precursors. Our goal was to conduct genome-wide CpG island methylation profiling to identify aberrantly methylated loci in IPMNs. EXPERIMENTAL DESIGN: We compared the CpG island methylation profiles of six IPMNs to normal primary pancreatic duct samples using methylation CpG island amplification (MCA) and Agilent CpG island microarray (MCAM) analysis. When selected 13 genes identified as differentially methylated by MCAM for methylation-specific PCR (MSP) analysis in an independent set of IPMNs and normal pancreas samples and conducted expression analysis of selected genes. RESULTS: We identified 2,259 loci as differentially methylated in at least one of six IPMNs including 245 genes hypermethylated in IPMNs with high-grade dysplasia compared with normal pancreatic duct samples. Eleven of 13 genes evaluated by MSP were more commonly methylated in 61 IPMNs than in 43 normal pancreas samples. Several genes (BNIP3, PTCHD2, SOX17, NXPH1, EBF3) were significantly more likely to be methylated in IPMNs with high-grade than with low-grade dysplasia. One gene, SOX17, showed loss of protein expression by immunohistochemistry in 22% (19 of 88) of IPMNs. The most specific marker, BNIP3, was not methylated in any IPMNs with low-grade dysplasia or in normal pancreas samples. CONCLUSIONS: IPMNs undergo extensive aberrant CpG island hypermethylation. The detection of genes selectively methylated in high-grade IPMNs such as BNIP3 may have use in the clinical evaluation of IPMNs.


Subject(s)
Adenocarcinoma, Mucinous/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Papillary/genetics , CpG Islands/genetics , Membrane Proteins/genetics , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Adenocarcinoma, Mucinous/metabolism , Adenocarcinoma, Mucinous/pathology , Aged , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Papillary/metabolism , Carcinoma, Papillary/pathology , DNA Methylation/genetics , Female , Genome-Wide Association Study , Humans , Immunohistochemistry , Male , Membrane Proteins/metabolism , Middle Aged , Oligonucleotide Array Sequence Analysis , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
10.
Clin Cancer Res ; 17(13): 4341-54, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21610144

ABSTRACT

PURPOSE: The goal of this study was to comprehensively identify CpG island methylation alterations between pancreatic cancers and normal pancreata and their associated gene expression alterations. EXPERIMENTAL DESIGN: We employed methylated CpG island amplification followed by CpG island microarray, a method previously validated for its accuracy and reproducibility, to analyze the methylation profile of 27,800 CpG islands covering 21 MB of the human genome in nine pairs of pancreatic cancer versus normal pancreatic epithelial tissues and in three matched pairs of pancreatic cancer versus lymphoid tissues from the same individual. RESULTS: This analysis identified 1,658 known loci that were commonly differentially methylated in pancreatic cancer compared with normal pancreas. By integrating the pancreatic DNA methylation status with the gene expression profiles of the same samples before and after treatment with the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine, and the histone deacetylase inhibitor, trichostatin A, we identified dozens of aberrantly methylated and differentially expressed genes in pancreatic cancers including a more comprehensive list of hypermethylated and silenced genes that have not been previously described as targets for aberrant methylation in cancer. CONCLUSION: We expected that the identification of aberrantly hypermethylated and silenced genes will have diagnostic, prognostic, and therapeutic applications.


Subject(s)
Adenocarcinoma/genetics , DNA Methylation/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genome-Wide Association Study , Pancreatic Neoplasms/genetics , Promoter Regions, Genetic , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Cell Adhesion/genetics , Cell Line, Tumor , CpG Islands/genetics , Epigenomics , Genetic Loci , Humans , Multigene Family/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pluripotent Stem Cells/metabolism , Signal Transduction/genetics , Wnt Proteins/metabolism
11.
Cancer Res ; 70(13): 5226-37, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20551052

ABSTRACT

Aberrant DNA methylation and microRNA expression play important roles in the pathogenesis of pancreatic cancer. While interrogating differentially methylated CpG islands in pancreatic cancer, we identified two members of miR-200 family, miR-200a and miR-200b, that were hypomethylated and overexpressed in pancreatic cancer. We also identified prevalent hypermethylation and silencing of one of their downstream targets, SIP1 (ZFHX1B, ZEB2), whose protein product suppresses E-cadherin expression and contributes to epithelial mesenchymal transition. In a panel of 23 pancreatic cell lines, we observed a reciprocal correlation between miR-200, SIP1, and E-cadherin expression, with pancreatic cancer-associated fibroblasts showing the opposite expression pattern to most pancreatic cancers. In Panc-1 cells, which express SIP1, have low E-cadherin expression, and do not express miR-200a or miR-200b, treatment with miR-200a and miR-200b downregulated SIP1 mRNA and increased E-cadherin expression. However, most pancreatic cancers express miR-200a and miR-200b, but this expression does not affect SIP1 expression, as the SIP1 promoter is silenced by hypermethylation and in these cancers E-cadherin is generally expressed. Both miR-200a and miR-200b were significantly elevated in the sera of pancreatic cancer and chronic pancreatitis patients compared with healthy controls (P < 0.0001), yielding receiver operating characteristic curve areas of 0.861 and 0.85, respectively. In conclusion, most pancreatic cancers display hypomethylation and overexpression of miR-200a and miR-200b, silencing of SIP1 by promoter methylation, and retention of E-cadherin expression. The elevated serum levels of miR-200a and miR-200b in most patients with pancreatic cancer could have diagnostic utility.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , DNA Methylation , MicroRNAs/genetics , Nerve Tissue Proteins/genetics , Pancreatic Neoplasms/genetics , RNA-Binding Proteins/genetics , Adult , Cadherins/biosynthesis , Cadherins/genetics , Carcinoma, Pancreatic Ductal/blood , Carcinoma, Pancreatic Ductal/metabolism , Case-Control Studies , Cell Line, Tumor , Female , Gene Silencing , Humans , Male , MicroRNAs/biosynthesis , MicroRNAs/blood , Middle Aged , Nerve Tissue Proteins/biosynthesis , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/metabolism , RNA-Binding Proteins/biosynthesis
12.
Mol Cancer Res ; 8(6): 821-32, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20530583

ABSTRACT

Genes that are differentially expressed in pancreatic cancers and under epigenetic regulation are of considerable biological and therapeutic interest. We used global gene expression profiling and epigenetic treatment of pancreatic cell lines including pancreatic cancer cell lines, pancreatic cancer-associated fibroblasts, and cell lines derived from nonneoplastic pancreata. We examined expression and epigenetic alterations of cyclooxygenase-1 (COX-1) and COX-2 in pancreatic cancers and normal pancreas and performed proliferation, knockdown, and coculture experiments to understand the role of stromal sources of prostaglandins for pancreatic cancers. We identify COX-1 as a gene under epigenetic regulation in pancreatic cancers. We find that COX-1 expression is absent in many pancreatic cancer cells and some of these cancers also lack COX-2 expression. Suspecting that such cancers must rely on exogenous sources of prostaglandins, we show that pancreatic cancer stromal cells, such as fibroblasts expressing COX-1 and COX-2, are a likely source of prostaglandins for pancreatic cancer cells deficient in COX. Knocking down the prostaglandin transporter multidrug resistance-associated protein-4 in fibroblasts suppresses the proliferation of cocultured pancreatic cancer cells lacking COX. Pancreatic cancers that lack COX can use exogenous sources of prostaglandins. Blocking multidrug resistance-associated protein-4 may be a useful therapeutic strategy to deplete COX-deficient pancreatic cancers of prostaglandins.


Subject(s)
Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/genetics , Prostaglandin-Endoperoxide Synthases/deficiency , Prostaglandin-Endoperoxide Synthases/genetics , Prostaglandins/physiology , Cell Communication/genetics , Cell Line, Tumor , Cyclooxygenase 1/deficiency , Cyclooxygenase 1/genetics , Cyclooxygenase 2/deficiency , Cyclooxygenase 2/genetics , Dinoprostone/physiology , Down-Regulation/genetics , Fibroblasts/enzymology , Fibroblasts/metabolism , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Multidrug Resistance-Associated Proteins/genetics , Pancreatic Neoplasms/metabolism , Prostaglandins/biosynthesis , Stromal Cells/enzymology , Stromal Cells/metabolism
13.
Clin Cancer Res ; 16(6): 1781-9, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20215540

ABSTRACT

PURPOSE: Accumulating evidence suggests that cancer-associated stromal fibroblasts (CAF) contribute to tumor growth by actively communicating with cancer cells. Our aim is to identify signaling pathways involved in tumor-stromal cell interactions in human pancreatic cancer. EXPERIMENTAL DESIGN: We established primary fibroblast cultures from human pancreatic adenocarcinomas and nonneoplastic pancreas tissues. To identify differentially expressed genes in CAFs, we did gene expression profiling of human pancreatic CAFs and nonneoplastic pancreatic fibroblasts. RESULTS: The Hedgehog receptor Smoothened (SMO) was upregulated in CAFs relative to control fibroblasts. CAFs expressing SMO could transduce the Sonic hedgehog signal to activate Gli1 expression, and small interfering RNA knockdown of SMO blocked the induction of Gli1 in these cells. Stromal fibroblasts of human primary pancreatic adenocarcinomas overexpressed Smo compared with normal pancreatic fibroblasts. CONCLUSIONS: These findings implicate overexpression of Smo as a mechanism for the activation of Hedgehog signaling in human pancreatic CAFs and suggest that stromal cells may be a therapeutic target for Smo antagonists in pancreatic cancer.


Subject(s)
Adenocarcinoma/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Fibroblasts/metabolism , Hedgehog Proteins/metabolism , Pancreatic Neoplasms/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line , Female , Fibroblasts/pathology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/genetics , Humans , Immunoenzyme Techniques , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Reverse Transcriptase Polymerase Chain Reaction , Smoothened Receptor , Stromal Cells/metabolism , Stromal Cells/pathology , Tissue Array Analysis , Transcription Factors/genetics , Transcription Factors/metabolism , Zinc Finger Protein GLI1
14.
Cancer Biol Ther ; 9(4): 321-9, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20234167

ABSTRACT

DNA methyltransferase I (DNMT1) is the major methyltransferase responsible for methylating DNA and is overexpressed in many cancers. DNMT1 is also a therapeutic target for chemotherapy and chemoprevention. We hypothesized that loss of DNMT1 copy number could result in reduced DNMT1 levels and greater sensitivity to DNMT1 inhibitors. We examined DNMT1 expression in pancreatic cancers by immunohistochemistry and western blotting. We also examined DNMT1 copy number in 20 pancreatic cancer cell lines using Affymetrix SNP arrays and correlated copy number with DNMT1 expression. We tested 8 pancreatic cancer cell lines with DNMT1 inhibitors and measured growth inhibition. We identified overexpression of DNMT1 relative to normal pancreatic duct in 78.7% of pancreatic cancers (37/47) by immunohistochemistry and in 16/20 pancreatic cancer cell lines by western blot. Pancreatic cancer cell lines with loss of DNMT1 alleles tended to have lower DNMT1 expression (3 of 9 cell lines) compared to those without DNMT1 copy number loss (1 of 11). 5-aza-deoxycytidine (5-Aza-dC) treatment (1-10 uM) depleted DNMT1 in 7 of 8 pancreatic cancer cell lines. Three of four pancreatic cancers cell lines with low/normal DNMT1 expression were sensitive to growth inhibition by low dose 5-Aza-dC (1 uM), whereas only 1 of 4 cell lines with high DNMT1 expression had growth inhibition, and this occurred without evidence of DNMT1 depletion suggesting a different mechanism for growth inhibition in this cell line. Loss of DNMT1 alleles may reduce DNMT1 levels in some pancreatic cancers. Pancreatic cancers with low DNMT1 expression tend to be more sensitive to low-dose 5-azadeoxycytidine.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , Pancreatic Neoplasms/metabolism , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Catechin/analogs & derivatives , Catechin/pharmacology , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Chromosomes, Human, Pair 19/genetics , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Decitabine , Drug Resistance, Neoplasm , Gene Dosage , Humans , Mutation , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , ras Proteins/genetics
15.
Gan To Kagaku Ryoho ; 37(2): 194-9, 2010 Feb.
Article in Japanese | MEDLINE | ID: mdl-20154472

ABSTRACT

Approximately 35-60% of patients with colorectal cancers will develop liver lesions during their life span. Previously colorectal liver metastases have traditionally been categorized as incurable systemic disease. However, the introduction of new chemotherapeutic regimens(e.g. FOLFOX or FOLFIRI)and recent technical innovations(e. g. staged hepatic resection and percutaneous transhepatic portal embolization)has allowed us to perform hepatic resection with curative intent. Additionally, a neoadjuvant strategy has expanded the criteria for liver resection, and more active molecular therapeutic agents are now available. As a result, a recent advancement has enhanced the overall 5-year survival from 30% to 58% for colorectal liver metastases. Despite these facts, many patients still experience a recurrence after hepatic resection. Modern treatment of colorectal liver metastases requires a multimodal approach to increase the number of patients who may benefit from surgical treatment of colorectal liver metastases.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Liver Neoplasms/diagnosis , Liver Neoplasms/drug therapy , Combined Modality Therapy , Hepatectomy , Humans , Liver Neoplasms/secondary , Liver Neoplasms/surgery , Survival Rate
16.
J Hepatobiliary Pancreat Sci ; 17(6): 745-51, 2010 Nov.
Article in English | MEDLINE | ID: mdl-19789835

ABSTRACT

BACKGROUND/PURPOSE: The Frey procedure, the coring out of the pancreatic head and longitudinal pancreaticojejunostomy, is a safe, easy, and reliable method to solve most of the problems associated with chronic pancreatitis. During long-term follow up, unexpected relapse in the pancreatic tail was encountered. The pattern of failure and the rationale for a new procedure to treat or prevent such relapse were investigated. METHODS: From 1992 to 2008, 71 patients with chronic pancreatitis underwent the Frey procedure at Tohoku University Hospital. The etiology was alcoholic in 92.6% of them, followed in incidence by idiopathic and hereditary chronic pancreatitis. In the primary operation, besides the Frey procedure, combined resection of the pancreatic tail was performed in three patients, and choledochoduodenostomy was performed in one patient. The follow-up rate was 92.9%, with a median period of 46 months. RESULTS: The incidence of early postoperative complications was 18.4%, with one reoperation for gastrointestinal bleeding from the splenic artery. Pain control was achieved in all patients and there was no operative mortality. During the long-term follow up of 62 patients with the Frey procedure, eight patients had relapse of inflammation and required reoperation. Five of these eight patients had a pseudocyst in the pancreatic tail and underwent distal pancreatectomy (DP). CONCLUSIONS: Relapse occurred in alcoholic middle-aged male patients, and in the patients with hereditary and idiopathic pancreatitis. Frey-DP and Frey-spleen-preserving DP (SPDP) procedures can be performed safely and effectively to treat the relapse and to prevent relapse in the pancreatic tail.


Subject(s)
Pancreatectomy/methods , Pancreaticojejunostomy/methods , Pancreatitis, Chronic/surgery , Adolescent , Adult , Aged , Female , Follow-Up Studies , Humans , Incidence , Japan/epidemiology , Male , Middle Aged , Postoperative Complications/epidemiology , Retrospective Studies , Secondary Prevention , Time Factors , Young Adult
17.
Cancer Epidemiol Biomarkers Prev ; 18(9): 2380-5, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19723916

ABSTRACT

Markers of early pancreatic cancer and its precursors are needed to improve the uniformly poor prognosis of this disease. Fatty acid synthase (FAS) catalyzes the synthesis of long-chain fatty acids and is overexpressed in most human solid tumors. We therefore evaluated serum FAS as a marker of pancreatic adenocarcinoma. FAS expression patterns in primary pancreatic adenocarcinomas, intraductal papillary mucinous neoplasms (IPMN), and chronic pancreatitis tissues were analyzed by immunohistochemistry. Serum FAS levels were determined by ELISA in 102 patients with pancreatic adenocarcinomas, in 42 patients with IPMNs, in 27 patients with chronic pancreatitis, and in 39 healthy control subjects. FAS protein was overexpressed in the ductal epithelium of 343 of 399 primary pancreatic adenocarcinomas (86.0%) and 28 of 30 IPMNs (93.3%), and in the islet and ductal cells in 3 of 54 chronic pancreatitis tissues (5.6%), whereas normal ductal epithelium lacked FAS expression. Serum FAS levels were significantly higher in patients with pancreatic ductal adenocarcinoma (first quartile median, 22.0; 4.5 ng/mL), in patients with IPMNs (20.7; 9.4 ng/mL), and in patients with chronic pancreatitis (31.1; 11.9 ng/mL) than in healthy controls (0; 0 ng/mL). FAS levels declined postoperatively in 8 of 9 patients with pancreatic adenocarcinoma and elevations of their preoperative serum FAS. In conclusion, serum FAS levels are elevated in patients with pancreatic cancer and IPMNs and are associated with neoplastic overexpression of FAS.


Subject(s)
Adenocarcinoma/enzymology , Biomarkers, Tumor/blood , Fatty Acid Synthases/blood , Pancreatic Neoplasms/enzymology , Adenocarcinoma/blood , Aged , Aged, 80 and over , Blotting, Western , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunohistochemistry , Male , Middle Aged , Pancreatic Neoplasms/blood
18.
Pancreatology ; 9(3): 293-301, 2009.
Article in English | MEDLINE | ID: mdl-19407485

ABSTRACT

Aberrant expression of microRNAs (miRNAs) has emerged as an important hallmark of cancer. However, the putative mechanisms regulating miRNAs per se are only partially known. It is well established that many tumor suppressor genes in human cancers are silenced by chromatin alterations, including promoter methylation and histone deacetylation. We postulated that miRNAs undergo similar epigenetic inactivation in pancreatic cancer. Two human pancreatic cancer cell lines - MiaPACA-2 and PANC-1 - were treated with the demethylating agent, 5-aza-2'-deoxycytidine (5-Aza-dC) or the histone deacetylase inhibitor, trichostatin A, as well as the combination of the two. Expression of miRNAs in control and treated cell lines was assessed using a custom microarray platform. Fourteen miRNAs were upregulated two-fold or greater in each of the cell lines following exposure to both chromatin-modifying agents, including 5 that were in common (miR-107, miR-103, miR-29a, miR-29b, and miR-320) to both MiaPACA-2 and PANC-1. The differential overexpression of miR-107 in the treated cancer cell lines was confirmed by Northern blot assays. Methylation-specific PCR assays for assessment of CpG island methylation status in the 5' promoter region of the miR-107 primary transcript demonstrated complete loss of methylation upon exposure to 5-Aza-dC. Enforced expression of miR-107 in MiaPACA-2 and PANC-1 cells downregulated in vitro growth, and this was associated with repression of the putative miR-107 target, cyclin-dependent kinase 6, thereby providing a functional basis for the epigenetic inactivation of this miRNA in pancreatic cancer.


Subject(s)
Cyclin-Dependent Kinase 6/genetics , Gene Silencing , MicroRNAs/genetics , Pancreatic Neoplasms/genetics , Conserved Sequence , DNA Primers , DNA, Neoplasm/genetics , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Oligonucleotide Array Sequence Analysis , Pancreatic Neoplasms/enzymology , Promoter Regions, Genetic , RNA, Neoplasm/genetics
19.
Cancer Epidemiol Biomarkers Prev ; 18(4): 1328-30, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19336541

ABSTRACT

It has been reported that germline mutations in the palladin gene (PALLD) cause the familial aggregation of pancreatic cancer, but the evidence is weak and controversial. We sequenced the coding regions of PALLD in 48 individuals with familial pancreatic cancer. We did not find any deleterious mutations and find no evidence to implicate mutations in PALLD as a cause of familial pancreatic cancer.


Subject(s)
Cytoskeletal Proteins/genetics , Genetic Predisposition to Disease , Pancreatic Neoplasms/genetics , Phosphoproteins/genetics , Polymorphism, Single Nucleotide/genetics , Baltimore/epidemiology , Genotype , Humans , Pancreatic Neoplasms/epidemiology , Prognosis
20.
Int J Clin Exp Pathol ; 2(4): 310-26, 2009.
Article in English | MEDLINE | ID: mdl-19158989

ABSTRACT

Pancreatic cancer remains a major therapeutic challenge. In 2008, there will be approximately 37,680 new cases and 34,290 deaths attributable to pancreatic cancer in the United States (U.S.), making it the fourth leading cause of cancer-related death. Recent comprehensive pancreatic cancer genome project found that pancreatic adenocarcinomas harbored 63 intragenic mutations or amplifications/homozygous deletions and these alterations clustered in 12 signaling pathways. In addition to widespread genetic alterations, it is now apparent that epigenetic mechanisms are also central to the evolution and progression of human cancers. Since epigenetic silencing processes are mitotically heritable, they can drive neoplastic progression and undergo the same selective pressure as genetic alterations. This review will describe recent developments in cancer epigenetics and their importance in our understanding of pancreatic adenocarcinomas.

SELECTION OF CITATIONS
SEARCH DETAIL
...