Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
2.
Endocr Pathol ; 24(2): 83-91, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23657967

ABSTRACT

The aim of this study was to assess the suitability of using real-time quantitative PCR (RT-qPCR) to characterize neuroendocrine (NE) tumors of the pancreas. For a series of tumors, we evaluated several genes of interest, and the data were matched with the "classical" immunohistochemical (IHC) features. In 21 cases, we extracted RNA from formalin-fixed paraffin-embedded (FFPE) blocks, and in nine cases, we also extracted RNA from fresh-frozen tissue. The RT-qPCR procedure was performed using two sets of customized arrays. The test using the first set, covering 96 genes of interest, was focused on assessing the feasibility of the procedure, and the results were used to select 18 genes indicative of NE differentiation, clinical behavior, and therapeutic responsiveness for use in the second set of arrays. Threshold cycle (Ct) values were used to calculate the fold-changes in gene expression using the 2-∆∆Ct method. Statistical procedures were used to analyze the results, which were matched with the IHC and follow-up data. Material from fresh-frozen samples performed better in terms of the level of amplification, but acceptable and concordant results were also obtained from FFPE samples. In addition, high concordance was observed between the mRNA and protein expression levels of somatostatin receptor type 2A (R = 0.52, p = 0.016). Genes associated with NE differentiation, as well as the gastrin-releasing peptide receptor and O-6-methylguanine-DNA methyltransferase genes, were underexpressed, whereas angiogenesis-associated markers (CDH13 and SLIT2) were overexpressed in tissues with malignant behavior. The RT-qPCR procedure is practical and feasible in economic terms for the characterization of NE tumors of the pancreas and can complement morphological and IHC-based evaluations. Thus, the results of the RT-qPCR procedure might offer an objective basis for therapeutic choices.


Subject(s)
Carcinoma, Islet Cell/pathology , Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms/pathology , Real-Time Polymerase Chain Reaction/methods , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Islet Cell/genetics , Carcinoma, Islet Cell/metabolism , DNA, Neoplasm/analysis , Female , Frozen Sections , Humans , Immunohistochemistry/methods , Male , Middle Aged , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , RNA, Messenger/metabolism , Reproducibility of Results
3.
Pathol Int ; 60(5): 358-64, 2010 May.
Article in English | MEDLINE | ID: mdl-20518885

ABSTRACT

Endoscopic ultrasound-guided fine-needle aspiration (EUS-FNA) has enabled clinicians to histologically diagnose pancreatic tumors. However, EUS-FNA specimens often result in tiny fragmented tissues, so auxiliary utilities are necessary. Using immunostaining of CK7, CDX2, neuroendocrine markers and KRAS mutation analysis, we examined 57 FNA cell block sections and 61 surgically-resected specimens (25 invasive ductal carcinomas, 25 endocrine tumors, and 11 acinar cell tumors). In the majority of the matched pairs, the diagnoses between EUS-FNA and surgical specimens were concordant using the following criteria: neuroendocrine markers negative, CK7 positive, and mutated KRAS gene for invasive ductal carcinomas; neuroendocrine markers diffusely positive, CK7 and CDX2 negative, and wild-type KRAS gene for well-differentiated endocrine tumors; and neuroendocrine markers no more than focal positive, CK7 and CDX2 with various staining patterns, and wild-type KRAS gene for acinar cell carcinomas. Expression of CK7 and/or CDX2 in addition to KRAS mutations were occasionally seen in endocrine carcinomas, but not in well-differentiated endocrine tumors, suggesting that ductal differentiation in an endocrine tumor may be a predictor of aggressive disease. The usefulness of these markers was confirmed using 13 additional pancreatic tumors, prospectively. Although minimal in selection, these markers are helpful in making diagnosis from EUS-FNA specimens of the major pancreatic tumors.


Subject(s)
Carcinoma, Acinar Cell/diagnosis , Carcinoma, Islet Cell/diagnosis , Carcinoma, Pancreatic Ductal/diagnosis , Endosonography/methods , Pancreatic Neoplasms/diagnosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Biopsy, Fine-Needle , CDX2 Transcription Factor , Carcinoma, Acinar Cell/genetics , Carcinoma, Acinar Cell/metabolism , Carcinoma, Islet Cell/genetics , Carcinoma, Islet Cell/metabolism , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , DNA Mutational Analysis , DNA, Neoplasm/analysis , Homeodomain Proteins/metabolism , Humans , Keratin-7/metabolism , Pancreas/metabolism , Pancreas/pathology , Pancreatectomy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Prognosis , Prospective Studies , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins p21(ras) , ras Proteins/genetics , ras Proteins/metabolism
4.
Cancer Res ; 70(5): 1759-65, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20160037

ABSTRACT

Junctional adhesion molecule-A (JAM-A)-null dendritic cells (DCs) are more motile and effective than their wild-type counterpart in promoting contact hypersensitivity reaction. Here, we show that the growth and aggressiveness of pancreatic islet cell carcinoma induced by SV40 T antigen expression in beta cells (Rip1Tag2 mice) are significantly reduced in JAM-A-null mice. Because these tumor cells do not express JAM-A, we focused on changes in stroma reactivity. In the absence of JAM-A, tumors showed a small but significant reduction in angiogenesis and a marked increase in the immune reaction with enhanced infiltration of DCs (CD11c+ and MHC-II+) and CD4+ and CD8+ lymphocytes. In contrast, phagocyte number was not affected. DC capacity to produce cytokines was not significantly altered, but transmigration through JAM-A-null endothelial cells was increased as compared with JAM-A-positive endothelium. On adoptive transfer, JAM-A(-/-) DCs were recruited to tumors at slightly but significantly higher rate than JAM-A(+/+) DCs. Ablation of CD4+ and CD8+ cells with specific antibodies abrogated the inhibitory effect of JAM-A deletion on tumor growth and angiogenesis. These findings support the idea that, in the Rip1Tag2 tumor model, abrogation of JAM-A reduces cancer development by increasing antitumor immune response.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Islet Cell/immunology , Cell Adhesion Molecules/deficiency , Dendritic Cells/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Pancreatic Neoplasms/immunology , Receptors, Cell Surface/deficiency , Animals , Carcinoma, Islet Cell/genetics , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Female , Male , Mice , Mice, Knockout , Pancreatic Neoplasms/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology
5.
Am J Surg Pathol ; 34(4): 510-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20182344

ABSTRACT

BACKGROUND: Pancreatic acinar cell carcinomas (ACCs) are clinically and pathologically distinct from pancreatic ductal adenocarcinomas (PDAs). Whereas endocrine differentiation has been well shown in ACCs, significant ductal components are rare. This paper reviews the clinicopathologic features of a series of ACCs with prominent ductal differentiation. DESIGN: Cases of pancreatic ACCs with significant ductal differentiation were identified in the surgical pathology databases of 2 academic centers. Patient clinical information, gross and histologic features, and histochemical and immunohistochemical (IHC) results were recorded. Cases were tested for KRAS2 mutations. RESULTS: Eleven cases were identified (10 men and 1 woman; age range 52 to 79 y). Four patients presented with jaundice. At last follow-up, 7 patients died of disease and 2 others had recurrences. Tumors measured between 2 and 5.5 cm and were ill-defined, nodular, and multilobulated. Ten were located in the head of the pancreas. All but 2 exhibited extrapancreatic invasion. All cases showed significant evidence of both acinar and ductal differentiation, estimated to be at least 25% of the neoplastic cells, and 3 cases in addition had endocrine differentiation in more than 25% of cells. Five cases were predominately acinar with intracellular and sometimes extracellular mucin ("mucinous acinar cell carcinoma" pattern). Six cases seemed more mixed with areas recapitulating typical PDAs whereas the other portions of the tumors seemed akin to typical acinar cell carcinomas ("combined acinar and ductal" pattern). IHC positive staining results were as: trypsin (92%), chymotrypsin (92%), monoclonal carcinoembryonic antigen (100%), CK19 (100%), B72.3 (73%), CA19.9 (73%), CD56 (18%), synaptophysin (36%), and chromogranin (36%). One case showed p53 over-expression aznd none showed DPC4/Smad4 loss. Two cases had KRAS2 mutations. CONCLUSION: Despite the early embryologic divergence of acinar and ductal cell lineages, rare pancreatic tumors have both acinar and ductal differentiation, usually predominantly the former. The clinical course is highly aggressive.


Subject(s)
Carcinoma, Acinar Cell/pathology , Carcinoma, Islet Cell/pathology , Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/pathology , Aged , Biomarkers, Tumor/metabolism , Carcinoma, Acinar Cell/genetics , Carcinoma, Acinar Cell/metabolism , Carcinoma, Acinar Cell/mortality , Carcinoma, Islet Cell/genetics , Carcinoma, Islet Cell/metabolism , Carcinoma, Islet Cell/mortality , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/mortality , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Combined Modality Therapy , DNA Mutational Analysis , DNA, Neoplasm/analysis , Female , Humans , Immunoenzyme Techniques , Male , Middle Aged , Mucins/metabolism , Mutation , Neoplasms, Multiple Primary , New York/epidemiology , Pancreatectomy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/mortality , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , Survival Rate , Virginia/epidemiology , ras Proteins/genetics
6.
J Clin Oncol ; 28(2): 245-55, 2010 Jan 10.
Article in English | MEDLINE | ID: mdl-19917848

ABSTRACT

PURPOSE: We investigated the global gene expression in a large panel of pancreatic endocrine tumors (PETs) aimed at identifying new potential targets for therapy and biomarkers to predict patient outcome. PATIENTS AND METHODS: Using a custom microarray, we analyzed 72 primary PETs, seven matched metastases, and 10 normal pancreatic samples. Relevant differentially expressed genes were validated by either quantitative real-time polymerase chain reaction or immunohistochemistry on tissue microarrays. RESULTS: Our data showed that: tuberous sclerosis 2 (TSC2) and phosphatase and tensin homolog (PTEN) were downregulated in most of the primary tumors, and their low expression was significantly associated with shorter disease-free and overall survival; somatostatin receptor 2 (SSTR2) was absent or very low in insulinomas compared with nonfunctioning tumors; and expression of fibroblast growth factor 13 (FGF13) gene was significantly associated with the occurrence of liver metastasis and shorter disease-free survival. TSC2 and PTEN are two key inhibitors of the Akt/mammalian target of rapamycin (mTOR) pathway and the specific inhibition of mTOR with rapamycin or RAD001 inhibited cell proliferation of PET cell lines. CONCLUSION: Our results strongly support a role for PI3K/Akt/mTOR pathway in PET, which ties in with the fact that mTOR inhibitors have reached phase III trials in neuroendocrine tumors. The finding of differential SSTR expression raises the potential for SSTR expression to be evaluated as a marker of response to somatostatin analogs. Finally, we identified FGF13 as a new prognostic marker that predicted poorer outcome in patients who were clinically considered free from disease.


Subject(s)
Carcinoma, Islet Cell/genetics , PTEN Phosphohydrolase/genetics , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins c-akt/genetics , Adolescent , Adult , Aged , Cell Line, Tumor , Down-Regulation , Female , Gene Expression Profiling , Humans , Male , Middle Aged , Neoplasm Metastasis , Signal Transduction , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics
7.
Mayo Clin Proc ; 84(9): 801-10, 2009 09.
Article in English | MEDLINE | ID: mdl-19720778

ABSTRACT

OBJECTIVE: To use fluorescence in situ hybridization (FISH) to visualize genetic abnormalities in interphase cell nuclei (interphase FISH) of acinar cell carcinoma, ductal adenocarcinoma, and islet cell carcinoma of the pancreas. PATIENTS AND METHODS: Between April 4, 2007, and December 4, 2008, interphase FISH was used to study paraffin-embedded preparations of tissue obtained from 18 patients listed in the Mayo Clinic Biospecimen Resource for Pancreas Research with a confirmed diagnosis of acinar cell carcinoma, ductal adenocarcinoma, islet cell carcinoma, or pancreas without evidence of neoplasia. FISH probes were used for chromosome loci of APC (see glossary at end of article for expansion of all gene symbols), BRCA2, CTNNB1, EGFR, ERBB2, CDKN2A, TP53, TYMP, and TYMS. These FISH probes were used with control probes to distinguish among various kinds of chromosome abnormalities of number and structure. RESULTS: FISH abnormalities were observed in 12 (80%) of 15 patients with pancreatic cancer: 5 of 5 patients with acinar cell carcinoma, 5 of 5 patients with ductal adenocarcinoma, and 2 (40%) of 5 patients with islet cell carcinoma. All 3 specimens of pancreatic tissue without neoplasia had normal FISH results. Gains of CTNNB1 due to trisomy 3 occurred in each tumor with acinar cell carcinoma but in none of the other tumors in this study. FISH abnormalities of all other cancer genes studied were observed in all forms of pancreatic tumors in this investigation. CONCLUSION: FISH abnormalities of CTNNB1 due to trisomy 3 were observed only in acinar cell carcinoma. FISH abnormalities of genes implicated in familial cancer, tumor progression, and the 5-fluorouracil pathway were common but were not associated with specific types of pancreatic cancer.


Subject(s)
Carcinoma, Acinar Cell/genetics , Carcinoma, Islet Cell/genetics , Carcinoma, Pancreatic Ductal/genetics , Chromosome Aberrations , Pancreatic Neoplasms/genetics , Carcinoma, Acinar Cell/pathology , Carcinoma, Islet Cell/pathology , Carcinoma, Pancreatic Ductal/pathology , Female , Humans , In Situ Hybridization, Fluorescence , Interphase/genetics , Male , Pancreatic Neoplasms/pathology
8.
Pancreas ; 38(8): 941-6, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19657311

ABSTRACT

OBJECTIVE: The goal of the study was to investigate the genetic and molecular basis of a novel syndrome of marked hyperglucagonemia and pancreatic alpha cell hyperplasia without glucagonoma syndrome. METHODS: The glucagon receptor (GCGR) gene and the glucagon gene were sequenced in a patient with hyperglucagonemia and pancreatic alpha cell hyperplasia without glucagonoma syndrome. Enhanced green fluorescent protein-conjugated wild type (WT) and mutant GCGR were used to characterize the functions of the mutant GCGR. RESULTS: The glucagon gene sequence was normal, but the GCGR sequencing uncovered a homozygous missense mutation, c.256C>T, p.P86S in the extracellular domain of GCGR. When expressed in human embryonic kidney 293 cells, GCGR P86S localized to the plasma membrane but bound 96% less radiolabeled glucagon than WT GCGR. The median effective concentration of glucagon-induced cyclic adenosine monophosphate production was 24 nmol/L for GCGR P86S but 2.4 nmol/L for WT GCGR. The patient's alpha cells also express glucagonlike peptide 1 and pancreatic polypeptide. CONCLUSIONS: We hereby report the first homozygous missense mutation in the human GCGR, which is associated with alpha cell hyperplasia and hyperglucagonemia. This mutation lowers the receptor's affinity to glucagon and decreases cyclic adenosine monophosphate production with physiological concentrations of glucagon. Thus, the P86S mutation in GCGR likely causes alpha cell hyperplasia and hyperglucagonemia.


Subject(s)
Carcinoma, Islet Cell/pathology , Glucagon-Secreting Cells/pathology , Glucagon/blood , Mutation, Missense , Pancreatic Neoplasms/pathology , Receptors, Glucagon/genetics , Aged , Amino Acid Sequence , Base Sequence , Carcinoma, Islet Cell/genetics , Carcinoma, Islet Cell/metabolism , Cell Line , Cell Membrane/metabolism , DNA Mutational Analysis , Female , Fluorescent Antibody Technique , Glucagon-Secreting Cells/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homozygote , Humans , Hyperplasia , Immunoblotting , Male , Molecular Sequence Data , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pedigree , Receptors, Glucagon/metabolism , Transfection
10.
Gastrointest Endosc ; 69(6): 1074-80, 2009 May.
Article in English | MEDLINE | ID: mdl-19152901

ABSTRACT

BACKGROUND: The clinical course of pancreatic endocrine tumors (PET) depends on tumor size, the presence of invasion or metastasis, the Ki-67 index, mitoses per high power field, and mutational damage. Most of this information is not available before surgery for clinical decision making or prognostication. OBJECTIVE: To evaluate PET EUS-guided FNA (EUS-FNA) microsatellite loss analysis in the context of PET-related mortality. DESIGN: A single institution retrospective cohort. PATIENTS: Patients with PET diagnosed by EUS-FNA who underwent DNA microsatellite loss analysis and at least 1 year of follow-up or subsequent death. INTERVENTION: PET microsatellite loss analysis results and current clinical status were compared. RESULTS: Twenty-nine patients were included in the final analysis; the mean age of the patients was 57 years, and 10 were women (35%). The mean follow-up was 33.7 months (median 30 months, range 2-66 months). Twelve patients had disease progression, and 8 died, all from disease-specific causes. Malignant PET contained multiple microsatellite losses, with a median fractional allelic loss (FAL) of 0.37 (range 0.12-0.69, interquartile range [IQR] 0.23-0.42), significantly different from benign PET, median FAL 0 (range 0-0.18, IQR 0-0.08, P < .0001). Survival analysis revealed a significant difference in disease recurrence or progression at 2 years (P < .0001) and in the 5-year survival between patients with FAL 0.2 (P < .0001). Logistic regression could not be performed because of the perfect association between an FAL >0.2 and disease status or mortality. LIMITATIONS: Retrospective design, referral bias, and DNA analysis availability. CONCLUSIONS: PET EUS-FNA microsatellite loss analysis provides preoperative prognostic information. An FAL >0.2 is not only associated with disease progression but also with mortality.


Subject(s)
Biopsy, Fine-Needle , Carcinoma, Islet Cell/diagnostic imaging , Carcinoma, Islet Cell/genetics , Endosonography , Loss of Heterozygosity/genetics , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/genetics , Ultrasonography, Interventional , Adult , Aged , Aged, 80 and over , Carcinoma, Islet Cell/mortality , Carcinoma, Islet Cell/pathology , Disease Progression , Female , Follow-Up Studies , Humans , Kaplan-Meier Estimate , Ki-67 Antigen/genetics , Male , Middle Aged , Neoplasm Staging , Pancreas/diagnostic imaging , Pancreas/pathology , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Prognosis
11.
Pancreas ; 38(1): 49-57, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19106744

ABSTRACT

OBJECTIVES: Homeobox gene caudal related homeobox gene 2 (CDX2) is an intestine-specific tumor suppressor gene. This study is intended to investigate the effect of CDX2 expression on cell proliferation and cyclin D1 expression in pancreatic cancer cells. METHODS: Four pancreatic ductal adenocarcinoma cell lines (PancQGO-1, BxPC-3, MIAPaCa-2, CFPAC-1), 1 islet carcinoma cell line (QGP-1), and 1 adenosquamous carcinoma cell line (KP-3) were analyzed for CDX1 and CDX2 expression using real-time reverse transcription-polymerase chain reaction and Western blot analysis. Proliferation of pancreatic cancer cells was analyzed using WST-1 assay after CDX2 transfection. Luciferase assay was performed to examine the effects of CDX2 on cyclin D1 transcriptional activity. RESULTS: CDX2 was expressed at a significantly higher level in QGP-1 cells than in KP-3 cells. Moreover, CDX2 was expressed at a middle level in 4 pancreatic ductal adenocarcinoma cells. Cell proliferation and cyclin D1 mRNA level were inhibited significantly after CDX2 transfection in pancreatic cancer cells. Furthermore, CDX2 inhibited exogenous nuclear factor kappaB-p65-induced luciferase gene expression in a dose-dependent manner. In addition, CDX2 inhibited pGL2HIVD1kappaB2-luciferase activity. CONCLUSIONS: CDX2 might play a role in inhibiting cell proliferation and repressing cyclin D1 transcriptional activity through the proximal nuclear factor kappaB binding site in pancreatic cancer cells.


Subject(s)
Cell Proliferation , Cyclin D1/genetics , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Pancreatic Neoplasms/genetics , Transcription, Genetic , CDX2 Transcription Factor , Carcinoma, Adenosquamous/genetics , Carcinoma, Adenosquamous/pathology , Carcinoma, Islet Cell/genetics , Carcinoma, Islet Cell/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cyclin D1/metabolism , Cyclooxygenase 2/genetics , Down-Regulation , Homeodomain Proteins/metabolism , Humans , NF-kappa B/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Promoter Regions, Genetic , RNA, Messenger/metabolism , Time Factors , Transfection
12.
Gastroenterology ; 134(5): 1544-54, 2008 May.
Article in English | MEDLINE | ID: mdl-18471525

ABSTRACT

BACKGROUND & AIMS: Cell adhesion molecule 1 (CADM1), mediates nerve-mast cell attachment and communication through homophilic binding. An immunohistochemical screen showed that CADM1 is expressed in pancreatic islets. Here, we determined the cell types expressing CADM1 and examined its role in nerve-islet cell interactions. METHODS: Immunohistochemistry and double-staining immunofluorescence were performed on murine and human pancreases and on islet cell tumors (ICTs). alphaTC6 cells, a murine alpha cell line, were cultured on neurite networks of superior cervical ganglia. Neurite-alphaTC6 cell attachment and communication were examined after nerves were activated specifically by scorpion venom. RESULTS: CADM1 was expressed on the plasma membrane in all 4 major types of islet cells, alpha, beta, D, and pancreatic polypeptide in human beings, but primarily in alpha cells in mice. In cocultures, alphaTC6 cell to neurite attachment was inhibited dose-dependently by an anti-CADM1 function-blocking antibody. In response to scorpion venom-evoked nerve activation, 36% of the alphaTC6 cells mobilized Ca(2+), and introduction of a CADM1-targeting small interfering RNA reduced the fraction of responding cells to 7%. In 21 human ICTs, CADM1 was present in the plasma membrane of 7, and the others were negative for CADM1. Six of the CADM1-expressing tumors were functional hormonally, whereas all but 2 of the CADM1-negative tumors were nonfunctional (P = .0032). CONCLUSIONS: CADM1 is a novel islet cell adhesion molecule mediating nerve-islet cell interactions. The strong correlation between CADM1 expression and hormonally functional phenotypes suggests that CADM1 is involved in hormone secretion from ICTs.


Subject(s)
Carcinoma, Islet Cell/metabolism , Gene Expression Regulation, Neoplastic , Immunoglobulins/genetics , Islets of Langerhans/metabolism , Membrane Proteins/genetics , Pancreatic Neoplasms/metabolism , RNA, Neoplasm/genetics , Tumor Suppressor Proteins/genetics , Adult , Animals , Carcinoma, Islet Cell/genetics , Carcinoma, Islet Cell/pathology , Cell Adhesion Molecule-1 , Cell Adhesion Molecules , Genes, Tumor Suppressor , Glucagon/metabolism , Humans , Immunoglobulins/biosynthesis , Immunohistochemistry , Islets of Langerhans/innervation , Islets of Langerhans/pathology , Membrane Proteins/biosynthesis , Mice , Neoplasms, Experimental , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Polymerase Chain Reaction , Tumor Suppressor Proteins/biosynthesis
13.
Hematol Oncol Clin North Am ; 21(3): 457-73; viii, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17548034

ABSTRACT

Most patients who have islet cell tumors, except those who have insulinomas, present with locally advanced or metastatic disease. In contrast with patients who have adenocarcinoma of the pancreas, those who have islet cell carcinomas can achieve long-term survival even if their disease is advanced. Liver-directed therapies, somatostatin analogs, and interferon are not curative but can be used to relieve tumor-associated symptoms. Similarly, palliative chemotherapy has been used with limited success. Advances in our understanding of the molecular mechanisms underlying tumor progression have translated into intense interest in biologically based strategies to treat this disease.


Subject(s)
Carcinoma, Islet Cell , Common Bile Duct Neoplasms , Neuroendocrine Tumors , Pancreatic Neoplasms , Carcinoma, Islet Cell/diagnosis , Carcinoma, Islet Cell/genetics , Carcinoma, Islet Cell/therapy , Common Bile Duct Neoplasms/diagnosis , Common Bile Duct Neoplasms/genetics , Common Bile Duct Neoplasms/therapy , Humans , Neuroendocrine Tumors/diagnosis , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/therapy , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/therapy , Risk Factors
14.
Virchows Arch ; 450(5): 549-57, 2007 May.
Article in English | MEDLINE | ID: mdl-17429687

ABSTRACT

Altered expression of recently described claudins (CLDNs) as members of tight junction (TJ) transmembrane proteins was noted in several malignancies. We aimed to analyze protein and messenger RNA (mRNA) expressions of different CLDNs in human pancreatic endocrine tumors (PET) and ductal adenocarcinomas. A total of 45 formalin-fixed, paraffin-embedded samples were studied. Immunohistochemistry and real-time reverse transcriptase polymerase chain reaction analysis were carried out for quantification of CLDN 1, -2, -3, -4, and -7 expressions. Normal acini and ducts showed strong CLDNs 1, -3, -4, and -7 and scattered CLDN 2 protein expressions, while Langerhans islands revealed only CLDN 3 and -7 expressions. CLDN 2 expression was found in the half of ductal adenocarcinomas, while the vast majority of endocrine tumors were negative. CLDN 1, -4, and -7 immunohistochemistry was positive in all adenocarcinomas, whereas endocrine tumors were completely negative for CLDNs 1 and -4. CLDN 3 and -7 proteins were detected in all endocrine tumors, while CLDN 3 in ductal adenocarcinomas was negative. The mRNA expression of CLDNs showed differences between endocrine tumors and ductal adenocarcinomas, similar as found for protein expression. Our findings support that PET and ductal carcinomas are specifically characterized by different expression pattern of CLDNs. High expressions of CLDN 3 in endocrine tumors and CLDN 4 in ductal carcinomas might attract them as targets for adjuvant therapy.


Subject(s)
Adenoma, Islet Cell/metabolism , Carcinoma, Islet Cell/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Membrane Proteins/metabolism , Pancreatic Neoplasms/metabolism , Adenoma, Islet Cell/genetics , Adenoma, Islet Cell/pathology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor , Carcinoma, Islet Cell/genetics , Carcinoma, Islet Cell/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/secondary , Female , Gene Expression , Humans , Immunoenzyme Techniques , Male , Membrane Proteins/genetics , Middle Aged , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , RNA, Messenger/metabolism , Retrospective Studies , Reverse Transcriptase Polymerase Chain Reaction
15.
Best Pract Res Clin Endocrinol Metab ; 21(1): 163-72, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17382271

ABSTRACT

Carcinoid and islet-cell carcinoma are often also known as low-grade neuroendocrine carcinomas. They are often slow-growing but can be resistant to standard therapy. While somatostatin analogues are often used to control hormonal syndromes, there is currently no therapy approved in the US for control of carcinoid tumor growth. For islet-cell carcinoma, streptozocin-based chemotherapy may induce tumor shrinkage, but second-line option are limited. This chapter reviews the molecular biology of neuroendocrine tumors, including the roles of MENIN, TSC2, NF-1, vHL, p53, bcl-2, bax, VEGF, IGF, PDGF, EGFR, and mTOR. Recently, there has been interest in developing molecularly targeted therapy for this group of diseases. Phase-II studies with imatinib, bevacizumab, sunitinib, gefitnib, temsirolimus, and everolimus (RAD001) have completed accrual. Encouraging results have been observed in studies with VEGF and mTOR inhibitors. Phase-III study of bevacizumab is planned in the US. Large-scale multinational phase-II and -III studies of everolimus are under way.


Subject(s)
Carcinoma, Islet Cell/therapy , Gene Targeting , Genetic Therapy , Neuroendocrine Tumors/therapy , Pancreatic Neoplasms/therapy , Biomarkers, Tumor/genetics , Carcinoma, Islet Cell/genetics , Genes, abl , Genes, erbB-1 , Humans , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/therapy , Neuroendocrine Tumors/genetics , Pancreas/blood supply , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/genetics , Protein Kinases/genetics , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-ret/genetics , Receptors, Platelet-Derived Growth Factor/genetics , TOR Serine-Threonine Kinases
16.
PLoS One ; 2(3): e323, 2007 Mar 28.
Article in English | MEDLINE | ID: mdl-17389914

ABSTRACT

BACKGROUND: Cancers of the pancreas originate from both the endocrine and exocrine elements of the organ, and represent a major cause of cancer-related death. This study provides a comprehensive assessment of gene expression for pancreatic tumors, the normal pancreas, and nonneoplastic pancreatic disease. METHODS/RESULTS: DNA microarrays were used to assess the gene expression for surgically derived pancreatic adenocarcinomas, islet cell tumors, and mesenchymal tumors. The addition of normal pancreata, isolated islets, isolated pancreatic ducts, and pancreatic adenocarcinoma cell lines enhanced subsequent analysis by increasing the diversity in gene expression profiles obtained. Exocrine, endocrine, and mesenchymal tumors displayed unique gene expression profiles. Similarities in gene expression support the pancreatic duct as the origin of adenocarcinomas. In addition, genes highly expressed in other cancers and associated with specific signal transduction pathways were also found in pancreatic tumors. CONCLUSION: The scope of the present work was enhanced by the inclusion of publicly available datasets that encompass a wide spectrum of human tissues and enabled the identification of candidate genes that may serve diagnostic and therapeutic goals.


Subject(s)
Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma/surgery , Artifacts , Axin Protein , Carcinoma, Islet Cell/genetics , Carcinoma, Islet Cell/pathology , Carcinoma, Islet Cell/surgery , Cell Line, Tumor , DNA, Neoplasm/genetics , Gene Expression Profiling , Genetic Variation , Humans , Multigene Family , Oligonucleotide Array Sequence Analysis , Pancreas/cytology , Pancreas/physiology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/surgery , RNA, Neoplasm/genetics , RNA, Neoplasm/isolation & purification , Reference Values , Repressor Proteins/genetics
18.
J Leukoc Biol ; 80(4): 669-76, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16793908

ABSTRACT

Many previous reports have demonstrated that systemic administration of endostatin (ES), a proteolytic cleavage product of collagen type XVIII and an endogenous angiogenesis inhibitor, represses tumor angiogenesis in different preclinical tumor models with varying efficacy. For example, systemic delivery of recombinant ES to rat insulin promoter 1 (Rip1)T-antigen 2 (Tag2)-transgenic mice, a mouse model of pancreatic beta-cell carcinogenesis, has repressed tumor angiogenesis efficiently and with it, tumor growth. Here, we report that the transgenic expression of ES in Rip1ES-transgenic mice only interferes moderately with tumor growth in Rip1Tag2;Rip1ES double-transgenic mice. Tumor incidence is not reduced by the local expression of ES, and tumor outgrowth and progression to tumor malignancy are only retarded slightly. A significant effect of local ES expression on tumor angiogenesis is only apparent during the early stages of tumor development, where less angiogenic hyperplastic lesions are observed. Although efficiently produced and secreted by transgenic beta cells, locally expressed ES appears to be sequestered in the microenvironment, and its systemic levels are not increased. The results indicate that the antiangiogenic functions of ES critically depend on the mode of delivery and the site of expression: although its systemic application represses tumor angiogenesis and tumor growth efficiently, locally expressed ES appears to be less effective, and hence, additional mechanisms of solubilization or activation of latent ES seem to be required. These results have important implications about the modes of delivery used in antiangiogenic, therapeutic strategies, which are based on the antiangiogenic activities of ES.


Subject(s)
Angiogenesis Inhibitors/genetics , Carcinoma, Islet Cell/genetics , Endostatins/genetics , Neoplasms, Experimental/genetics , Neovascularization, Pathologic/genetics , Pancreas/pathology , Pancreatic Neoplasms/genetics , Angiogenesis Inhibitors/biosynthesis , Angiogenesis Inhibitors/pharmacology , Animals , Antigens, Polyomavirus Transforming/genetics , Carcinoma, Islet Cell/blood supply , Carcinoma, Islet Cell/metabolism , Cell Proliferation , Crosses, Genetic , Disease Models, Animal , Disease Progression , Endostatins/biosynthesis , Endostatins/pharmacology , Female , Humans , Insulin/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/metabolism , Neovascularization, Pathologic/pathology , Pancreas/blood supply , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/metabolism , Rats , Transgenes
19.
Pancreas ; 33(1): 38-44, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16804411

ABSTRACT

OBJECTIVES: To investigate the prevalence of PTCH (patched gene) and Smo (smoothened gene) expression in human pancreatic cancerous tissues and its association with clinical characteristics. METHODS: A rabbit polyclonal antibody against PTCH was prepared through the immunization of prokaryotic recombinant PTCH1170-1433 protein. The PTCH and Smo expression in 39 resected pancreas specimens from 28 patients with pancreatic cancer, 6 with chronic pancreatitis (as control), and 5 with pancreatic pseudocyst (as control) were detected by reverse transcriptase polymerase chain reaction and immunohistochemistry. The relationships between their expressions and pathological characteristics such as tumor sizes, degree of differentiation, nodal status, distant metastasis, and the blood sugar level were analyzed. RESULTS: The prevalence of PTCH and Smo expressions in cancerous tissues were 71.4% (20/28) and 53.6% (15/28), respectively, whereas no expression in the nontumor pancreas tissues was found. Both PTCH and Smo expressions correlated with the low levels of tumor tissue differentiation (P < 0.05) and PTCH and Smo expressions in islet cells of cancerous tissues associated with hyperglycemia. CONCLUSIONS: Because aberrant expressions of PTCH and Smo were common in human pancreatic carcinoma tissues and were associated with the low-level differentiation of tumor tissue and hyperglycemia, this indicated that these molecules played a fundamental role in pancreas tumorigenesis and were regarded as new targets for diagnosis and treatment of human pancreatic cancer.


Subject(s)
Carcinoma, Islet Cell/genetics , Carcinoma, Pancreatic Ductal/genetics , Gene Expression Regulation, Neoplastic , Hyperglycemia/metabolism , Pancreatic Neoplasms/genetics , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled/metabolism , Adult , Aged , Carcinoma, Islet Cell/metabolism , Carcinoma, Islet Cell/pathology , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Differentiation , Female , Humans , Hyperglycemia/genetics , Hyperglycemia/pathology , Immunohistochemistry , Male , Middle Aged , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Patched Receptors , Patched-1 Receptor , Receptors, Cell Surface/genetics , Receptors, G-Protein-Coupled/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Smoothened Receptor
20.
Oncol Rep ; 15(5): 1249-56, 2006 May.
Article in English | MEDLINE | ID: mdl-16596194

ABSTRACT

The activation of the insulin-like growth factor 1/IGF1 receptor system (IGF1/IGF1R) is a critical event in the transformation and tumorigenicity processes in a wide variety of human tumors. The IGF1/IGF1R system has been recently studied in carcinoid tumors that often arise in the gastrointestinal tract; these tumors are characterized by hypersecretion of bioamines and neuropeptides, leading to functional tumor disease. Two alternatively spliced IGF1R mRNA transcripts have been described to differ by only three nucleotides (CAG) in the coding sequence, resulting in an amino-acid change from the originally described Thr-Gly to an Arg in the extracellular portion of the receptor beta subunit. In transfected Chinese hamster ovary cells, the form without CAG (CAG-) exhibited an approximate 2-fold increase in IGF1 stimulation of activities required for its mitogenic properties. In this study, we examine the relative expression of the two IGF1R mRNA isoforms by a semiquantitative RT-PCR approach using highly standardized conditions, beta-2 microglobulin (B2M) as a reference gene and gel imaging analysis. We analyzed a large series of human neuroendocrine tumors (32 samples) and 9 normal tissues. A significant higher expression of both isoforms in the tumor samples (approximately 2-fold increase) was found, while a constant CAG+/CAG- IGF1R mRNA isoforms of an approximate 3:1 ratio was observed in all tumoral and normal cell types studied. The phylogenetic study of the IGF1R locus in several species suggests that human IGF1R CAG- mRNA isoform is evolutionarily more recent compared to the IGF1R CAG+ mRNA isoform and it could be used by the splicing apparatus at this intron/exon junction with a lower efficiency. This study highlights the relevance of IGF1R mRNA expression in neuroendocrine tumor cells, and the constant presence of 'subtle' alternative splicing for the IGF1R locus.


Subject(s)
Alternative Splicing , Neuroendocrine Tumors/genetics , RNA, Messenger/metabolism , Receptor, IGF Type 1/genetics , Adult , Aged , Amino Acid Sequence , Carcinoma, Islet Cell/genetics , Carcinoma, Islet Cell/metabolism , Carcinoma, Islet Cell/pathology , Cell Differentiation , DNA Primers/chemistry , Female , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Male , Middle Aged , Molecular Sequence Data , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Protein Isoforms , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Receptor, IGF Type 1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...