Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(8)2024 Apr 13.
Article in English | MEDLINE | ID: mdl-38673897

ABSTRACT

Pancreatic cancer, most frequently as ductal adenocarcinoma (PDAC), is the third leading cause of cancer death. Clear-cell primary adenocarcinoma of the pancreas (CCCP) is a rare, aggressive, still poorly characterized subtype of PDAC. We report here a case of a 65-year-old male presenting with pancreatic neoplasia. A histochemical examination of the tumor showed large cells with clear and abundant intracytoplasmic vacuoles. The clear-cell foamy appearance was not related to the hyperproduction of mucins. Ultrastructural characterization with transmission electron microscopy revealed the massive presence of mitochondria in the clear-cell cytoplasm. The mitochondria showed disordered cristae and various degrees of loss of structural integrity. Immunohistochemistry staining for NADH dehydrogenase [ubiquinone] 1 alpha subcomplex, 4-like 2 (NDUFA4L2) proved specifically negative for the clear-cell tumor. Our ultrastructural and molecular data indicate that the clear-cell nature in CCCP is linked to the accumulation of disrupted mitochondria. We propose that this may impact on the origin and progression of this PDAC subtype.


Subject(s)
Mitochondria , Pancreatic Neoplasms , Humans , Male , Aged , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/ultrastructure , Pancreatic Neoplasms/metabolism , Mitochondria/ultrastructure , Mitochondria/metabolism , Mitochondria/pathology , Adenocarcinoma, Clear Cell/pathology , Adenocarcinoma, Clear Cell/ultrastructure , Adenocarcinoma, Clear Cell/metabolism , Microscopy, Electron, Transmission , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/ultrastructure , Carcinoma, Pancreatic Ductal/metabolism , Immunohistochemistry
2.
Autophagy ; 16(3): 486-500, 2020 03.
Article in English | MEDLINE | ID: mdl-31177911

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive and lethal malignancies. Given that macroautophagy/autophagy activation is prevalent in PDAC, the dual roles of autophagy could be involved in PDAC heterogeneity. In this work, we demonstrated that TGFB1 induced autophagic flux through SMAD4-dependent or SMAD4-independent pathways based on a distinct genetic context. In SMAD4-positive PDAC cells, TGFB1-induced autophagy promoted proliferation and inhibited migration by decreasing the nuclear translocation of SMAD4. Conversely, TGFB1-induced autophagy inhibited proliferation and promoted migration in SMAD4-negative cells through the regulation of MAPK/ERK activation. TGFB1 expression also positively correlated with LC3B expression in PDAC specimens. A high level of LC3B was associated with unfavorable overall survival (OS) and disease-free survival (DFS) in SMAD4-negative PDAC patients, although LC3B could not predict OS and DFS for the 110 PDAC patients. Thus, TGFB1-induced autophagy contributed to the different patterns of PDAC progression. This knowledge can aid in improving our understanding of the molecular classification of PDAC and might guide the development of therapeutic strategies for PDAC, especially for SMAD4-negative PDAC.Abbreviations: CDH1: cadherin 1; CDH2: cadherin 2; CI: combination index; CQ: chloroquine; DFS: disease-free survival; EMT: epithelial-to-mesenchymal transition; ERK: extracellular signal-regulated protein kinase; GFP: green fluorescent protein; IHC: immunohistochemistry; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MAPK: mitogen-activated protein kinase; OS: overall survival; PBS: phosphate-buffered saline; PDAC: pancreatic ductal adenocarcinoma; RAP: rapamycin; RFP: red fluorescent protein; RT: room temperature; shRNA: short-hairpin RNA; SQSTM1: sequestosome 1; TCGA: The Cancer Genome Atlas; TEM: transmission electron microscopy; TGFB1: transforming growth factor beta 1; TMA: tissue microarray.


Subject(s)
Autophagy , Disease Progression , Pancreatic Neoplasms/pathology , Smad4 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma/ultrastructure , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/ultrastructure , Cell Line, Tumor , Cell Nucleus/metabolism , Female , Humans , MAP Kinase Signaling System , Male , Microtubule-Associated Proteins/metabolism , Middle Aged , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/ultrastructure , Prognosis , Sequestosome-1 Protein/metabolism , Pancreatic Neoplasms
3.
Pancreas ; 48(9): 1225-1231, 2019 10.
Article in English | MEDLINE | ID: mdl-31593010

ABSTRACT

OBJECTIVE: Pancreatic ductal adenocarcinoma (PDAC) is the third most common cause of cancer death in the United States. Improved characterized models of PDAC are needed for drug screening. METHODS: We grew 4 established pancreatic cancer cell lines in hanging drop cultures to produce spheroids. We also grew organoids from explanted xenografted PDAC and surgically resected primary PDAC. We performed transmission and scanning electron microscopy and compared findings with those of the normal pancreatic duct. We also performed single-cell cloning to determine the potential options for differentiation. RESULTS: Spheroids contained tight junctions and desmosomes but lacked zymogen granules, as expected. The former features were present in normal pancreatic duct but absent from PDAC cell lines grown in standard 2-dimensional culture. Spheroids functionally excluded macromolecules in whole mounts. Cells on the surface of PDAC spheroids were carpeted by microvilli except for rare cells with prominent stereocilia. Carpets of microvilli were also seen in low passage organoids produced from xenografts and surgically resected human PDAC, in addition to normal human pancreatic duct. We performed single-cell cloning and resulting spheroids produced both cell phenotypes at the same approximate ratios as those from bulk cultures. CONCLUSIONS: Pancreatic cancer spheroids/organoids are capable of biphenotypic differentiation.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Cell Culture Techniques/methods , Organoids/pathology , Pancreatic Ducts/pathology , Pancreatic Neoplasms/pathology , Spheroids, Cellular/pathology , Animals , Carcinoma, Pancreatic Ductal/ultrastructure , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Desmosomes/ultrastructure , Female , Heterografts/pathology , Heterografts/ultrastructure , Humans , Mice, Nude , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Organoids/ultrastructure , Pancreatic Ducts/ultrastructure , Pancreatic Neoplasms/ultrastructure , Spheroids, Cellular/ultrastructure , Tight Junctions/ultrastructure
4.
Biomaterials ; 192: 590-600, 2019 02.
Article in English | MEDLINE | ID: mdl-30553134

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a destructive cancer with poor prognosis. Both novel therapeutic targets and approaches are needed to improve the overall survival of PDAC patients. MicroRNA-212 (miR-212) has been reported as a tumor suppressor in multiple cancers, but its definitive role and exact mechanism in the progression of pancreatic cancer is unclear. In this study, we developed a new chimeric peptide (PL-1) composed of plectin-1-targeted PDAC-specific and arginine-rich RNA-binding motifs which could condense miRNA to self-assemble supramolecular nanoparticles. These nanoparticles could deliver miR-212 into PDAC cells specifically and efficiently which also showed good stability in RNase and serum. Moreover, we demonstrated that PL-1/miR-212 nanoparticles could dramatically enhance the chemotherapeutic effect of doxorubicin for PDAC both in vitro and in vivo. In terms of mechanism, combined miR-212 intervention by PL-1/miR-212 nanoparticles resulted in obvious decrease of USP9X expression (ubiquitin specific peptidase 9, X-linked, USP9X) and eventually enhanced the doxorubicin induced apoptosis and autophagy of PDAC cells. These findings provide a new promising anti-cancer strategy via PL-1/miR-212 nanoparticles and identify miR-212/USP9X as a new potential target for future systemic therapy against human PDAC.


Subject(s)
Adenocarcinoma/drug therapy , Carcinoma, Pancreatic Ductal/drug therapy , Doxorubicin/therapeutic use , MicroRNAs/administration & dosage , Nanoparticles/chemistry , Pancreatic Neoplasms/drug therapy , Peptides/chemistry , Adenocarcinoma/pathology , Adenocarcinoma/ultrastructure , Amino Acid Sequence , Animals , Apoptosis/drug effects , Autophagy/drug effects , Base Sequence , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/ultrastructure , Cell Line, Tumor , Doxorubicin/pharmacology , Humans , Male , Mice, Nude , MicroRNAs/genetics , Nanoparticles/ultrastructure , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/ultrastructure , Ubiquitin Thiolesterase/metabolism
5.
PLoS One ; 12(9): e0184451, 2017.
Article in English | MEDLINE | ID: mdl-28957348

ABSTRACT

Here we describe isolation and characterization of macrophage-tumor cell fusions (MTFs) from the blood of pancreatic ductal adenocarcinoma (PDAC) patients. The MTFs were generally aneuploidy, and immunophenotypic characterizations showed that the MTFs express markers characteristic of PDAC and stem cells, as well as M2-polarized macrophages. Single cell RNASeq analyses showed that the MTFs express many transcripts implicated in cancer progression, LINE1 retrotransposons, and very high levels of several long non-coding transcripts involved in metastasis (such as MALAT1). When cultured MTFs were transplanted orthotopically into mouse pancreas, they grew as obvious well-differentiated islands of cells, but they also disseminated widely throughout multiple tissues in "stealth" fashion. They were found distributed throughout multiple organs at 4, 8, or 12 weeks after transplantation (including liver, spleen, lung), occurring as single cells or small groups of cells, without formation of obvious tumors or any apparent progression over the 4 to 12 week period. We suggest that MTFs form continually during PDAC development, and that they disseminate early in cancer progression, forming "niches" at distant sites for subsequent colonization by metastasis-initiating cells.


Subject(s)
Carcinoma, Pancreatic Ductal/blood , Carcinoma, Pancreatic Ductal/pathology , Macrophages/pathology , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/pathology , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/ultrastructure , Cell Fusion , Cell Nucleus/pathology , Humans , Imaging, Three-Dimensional , Immunohistochemistry , Immunophenotyping , Male , Mice, Nude , Microscopy, Confocal , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/ultrastructure , Ploidies , Sequence Analysis, RNA , Single-Cell Analysis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Pancreatic Neoplasms
6.
Sci Rep ; 7: 42339, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28198398

ABSTRACT

The cancer drug gemcitabine (GEM) is a key drug for treating pancreatic ductal adenocarcinoma (PDAC), but PDAC cells develop chemoresistance after long-term administration. Since the tolerance was immediately spread to every PDAC tissue in a patient, it is assumed that some certain efficient mechanisms underlay in the development of chemoresistance. Changes in the levels of particular microRNAs or alterations in intercellular communication play a dominant role in chemoresistance development, and recent data also suggest that exosomes play an important role in this process. In this study, we revealed that the loop conferred chemoresistance in PDAC cells. The loop was as follows; 1, The long-term exposure of GEM increased miR-155 expression in PDAC cells. 2, The increase of miR-155 induced two different functions; exosome secretion and chemoresistance ability via facilitating the anti-apoptotic activity. 3, Exosome deliver the miR-155 into the other PDAC cells and induce the following function. The target therapy to miR-155 or the exosome secretion effectively attenuated the chemoresistance, and these results were validated with both clinical samples and in vivo experiments. This mechanism represents a novel therapeutic target in GEM treatment to PDAC.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm , Exosomes/metabolism , MicroRNAs/metabolism , Pancreatic Neoplasms/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma/ultrastructure , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/ultrastructure , Cell Line, Tumor , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Exosomes/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , MicroRNAs/genetics , Microdissection , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/ultrastructure , Prognosis , Time Factors , Up-Regulation/drug effects , Up-Regulation/genetics , Xenograft Model Antitumor Assays , Gemcitabine , Pancreatic Neoplasms
7.
World J Gastroenterol ; 22(18): 4466-83, 2016 May 14.
Article in English | MEDLINE | ID: mdl-27182158

ABSTRACT

AIM: To analyze the effect of three-dimensional (3D)-arrangement on the expression of epithelial-to-mesenchymal transition markers in pancreatic adenocarcinoma (PDAC) cells. METHODS: HPAF-II, HPAC, and PL45 PDAC cells were cultured in either 2D-monolayers or 3D-spheroids. Ultrastructure was analyzed by transmission electron microscopy. The expression of E-cadherin, ß-catenin, N-cadherin, collagen type I (COL-I), vimentin, α-smooth muscle actin (αSMA), and podoplanin was assayed by confocal microscopy in cells cultured on 12-mm diameter round coverslips and in 3D-spheroids. Gene expression for E-cadherin, Snail, Slug, Twist, Zeb1, and Zeb2 was quantified by real-time PCR. E-cadherin protein level and its electrophoretic pattern were studied by Western blot in cell lysates obtained from cells grown in 2D-monolayers and 3D-spheroids. RESULTS: The E-cadherin/ß-catenin complex was expressed in a similar way in plasma membrane cell boundaries in both 2D-monolayers and 3D-spheroids. E-cadherin increased in lysates obtained from 3D-spheroids, while cleavage fragments were more evident in 2D-monolayers. N-cadherin expression was observed in very few PDAC cells grown in 2D-monolayers, but was more evident in 3D-spheroids. Some cells expressing COL-I were observed in 3D-spheroids. Podoplanin, expressed in collectively migrating cells, and αSMA were similarly expressed in both experimental conditions. The concomitant maintenance of the E-cadherin/ß-catenin complex at cell boundaries supports the hypothesis of a collective migration for these cells, which is consistent with podoplanin expression. CONCLUSION: We show that a 3D-cell culture model could provide deeper insight into understanding the biology of PDAC and allow for the detection of marked differences in the phenotype of PDAC cells grown in 3D-spheroids.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Epithelial-Mesenchymal Transition , Pancreatic Neoplasms/pathology , Antigens, CD , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Cadherins/genetics , Cadherins/metabolism , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/ultrastructure , Cell Line, Tumor , Cell Shape , Cell Survival , Gene Expression Regulation, Neoplastic , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Microscopy, Confocal , Microscopy, Electron, Transmission , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/ultrastructure , Phenotype , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Spheroids, Cellular , beta Catenin/genetics , beta Catenin/metabolism
8.
Rom J Morphol Embryol ; 56(1): 207-22, 2015.
Article in English | MEDLINE | ID: mdl-25826507

ABSTRACT

In this study, we focus our interest on some peculiar infrastructural abnormalities detected in a pancreatic cancer case. Our electron microscopic observations underline the high plasticity of the pancreatic parenchyma cells. Tumor pancreatic exocrine lesions are represented by putative ductal and acinar cells, which proliferate and grow in a haphazard pattern, detrimental to endocrine counterpart. The tumor cells do not exhibit neither a pure ductular or ductal nor a pure acinar phenotype, but tumor lesions represented by neoplastic ductal cells with invasive growth are by far prevalently. In our pancreatic cancer case, electron microscopic investigation clearly shows that a plethora of the epithelial cells from the tumor lesions contain large areas of autophagy leading to the pleomorphic inclusions represented by fibrillary÷filamentous inclusions frequently associated with hyaline-amorphous material, and secondary lysosomes. One of the mostly striking and important finding in this report for a case of pancreatic cancer is the high fragility (extensive dissolutions) of plasma membrane of tumor cells leading to pseudo-syncytia formation. Desmosomal junctions are severely altered, almost missing. Plasma membranes showed shedding membrane vesicles. Extravasated inflammatory cells contribute to the dramatic and extensive destructive areas of epithelial cells as well as tumor-stroma counterpart, including the basement membrane. All above severe infrastructural abnormalities, especially down regulation of cell-cell and cell-extracellular matrix adhesions might result in aberrant cell behavior and, consequently, much care should be taken for the postoperatory patient evolution.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/pathology , Aged , Carcinoma, Pancreatic Ductal/ultrastructure , Cell Membrane/metabolism , Desmosomes/metabolism , Female , Humans , Inflammation , Lysosomes/metabolism , Microscopy, Electron, Transmission , Neoplasm Invasiveness , Pancreas/pathology , Pancreas/ultrastructure , Pancreatic Neoplasms/ultrastructure , Phenotype
9.
Pancreas ; 43(8): 1256-63, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25072283

ABSTRACT

OBJECTIVES: A functional vacuolar adenosine triphosphatase (v-ATPase) complex regulates canonical Wnt/ß-catenin signaling. The goal of this study was to identify the distribution of the v-ATPase in human and murine models of pancreatic intraepithelial neoplasms (PanINs) and assess its role in Wnt/ß-catenin signaling. METHODS: We evaluated the immunolabeling pattern of the v-ATPase in human PanIN specimens and murine PanIN-1 and PanIN-2 lesions obtained from Ptf1a(Cre/+); LSL-Kras(G12D) mice. Wnt/ß-catenin signaling was interrogated in primary PanIN cells by examining the phosphorylated levels of its surface coreceptor, low-density lipoprotein receptor-related protein-6 (LRP6), and its intracellular effector, nonphosphorylated ß-catenin. The response of primary PanIN cells to epidermal growth factor (EGF) was assessed in the absence and presence of the v-ATPase inhibitor, concanamycin. RESULTS: In advanced (PanIN-2), but not early (PanIN-1), lesions, the v-ATPase assumed a polarized phenotype. Blocking the v-ATPase disrupted Wnt/ß-catenin signaling in primary PanIN cells despite significantly higher levels of the total and activated Wnt cell surface coreceptor, LRP6. Vacuolar adenosine triphosphatase blockade significantly decreased the total and activated levels of EGF receptor, a determinant of PanIN progression. The activation of EGF receptor and its intracellular mediator, p44/42 mitogen-activated protein kinase, was also reduced by v-ATPase blockade. This led to diminished proliferation in response to EGF ligand. CONCLUSIONS: The v-ATPase regulates Wnt/ß-catenin and EGF receptor signaling in PanINs.


Subject(s)
Carcinoma, Pancreatic Ductal/enzymology , Membrane Proteins/analysis , Neoplasm Proteins/analysis , Pancreatic Neoplasms/enzymology , Vacuolar Proton-Translocating ATPases/analysis , Wnt Signaling Pathway/physiology , Adenocarcinoma in Situ/enzymology , Adenocarcinoma in Situ/ultrastructure , Alcian Blue , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/ultrastructure , Cell Line, Tumor , Cell Polarity , Disease Progression , Epidermal Growth Factor/pharmacology , ErbB Receptors/analysis , ErbB Receptors/drug effects , Humans , Islets of Langerhans/enzymology , Islets of Langerhans/ultrastructure , Low Density Lipoprotein Receptor-Related Protein-6/analysis , Mice , Mice, Mutant Strains , Mitogen-Activated Protein Kinase 3/analysis , Mitogen-Activated Protein Kinase 3/physiology , Neoplasm Grading , Neoplasm Proteins/physiology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/ultrastructure , Protein Transport , Staining and Labeling , Vacuolar Proton-Translocating ATPases/physiology
10.
Ultrastruct Pathol ; 37(3): 227-31, 2013 May.
Article in English | MEDLINE | ID: mdl-23650995

ABSTRACT

The aim of our study was to analyse the mitochondrial ultrastructure in primary ductal adenocarcinomas of the pancreas and to compare it with normal pancreatic cells. 52 samples of adenocarcinoma of the pancreas obtained by surgical resection or by endosonographic biopsy were examined. Compared to normal mitochondrial ultrastructure in non-tumorous cells, the mitochondria in cancer cells had a dense matrix and condensed configuration or with lucent-swelling matrix associated with disarrangement and distortion of cristae and partial or total cristolysis. Functionally, these structural alterations presume the presence of hypoxia-tolerant and hypoxia-sensitive cancer cells.


Subject(s)
Carcinoma, Pancreatic Ductal/ultrastructure , Mitochondria/ultrastructure , Pancreatic Neoplasms/ultrastructure , Biopsy , Endosonography , Humans , Microscopy, Electron
11.
J Pathol ; 227(3): 325-35, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22262369

ABSTRACT

Involvement of dysregulated autophagy in cancer growth and progression has been shown in different tumour entities, including pancreatic ductal adenocarcinoma (PDA). PDA is an extremely aggressive tumour characterized by a small population of highly therapy-resistant cancer stem cells (CSCs) capable of self-renewal and migration. We examined whether autophagy might be involved in the survival of CSCs despite nutrition and oxygen deprivation typical for the hypoxic tumour microenvironment of PDA. Immunohistochemistry revealed that markers for hypoxia, CSCs and autophagy are co-expressed in patient-derived tissue of PDA. Hypoxia starvation (H/S) enhanced clonogenic survival and migration of established pancreatic cancer cells with stem-like properties (CSC(high)), while pancreatic tumour cells with fewer stem cell markers (CSC(low)) did not survive these conditions. Electron microscopy revealed more advanced autophagic vesicles in CSC(high) cells, which exhibited higher expression of autophagy-related genes under normoxic conditions and relative to CSC(low) cells, as found by RT-PCR and western blot analysis. LC3 was already fully converted to the active LC3-II form in both cell lines, as evaluated by western blot and detection of accumulated GFP-LC3 protein by fluorescence microscopy. H/S increased formation of autophagic and acid vesicles, as well as expression of autophagy-related genes, to a higher extent in CSC(high) cells. Modulation of autophagy by inhibitors and activators resensitized CSC(high) to apoptosis and diminished clonogenicity, spheroid formation, expression of CSC-related genes, migratory activity and tumourigenicity in mice. Our data suggest that enhanced autophagy levels may enable survival of CSC(high) cells under H/S. Interference with autophagy-activating or -inhibiting drugs disturbs the fine-tuned physiological balance of enhanced autophagy in CSC and switches survival signalling to suicide.


Subject(s)
Autophagy , Carcinoma, Pancreatic Ductal/pathology , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/pathology , Tumor Microenvironment , Animals , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Autophagy/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/ultrastructure , Cell Hypoxia , Cell Line, Tumor , Cell Movement , Cell Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Mice , Mice, Nude , Microscopy, Electron , Microscopy, Fluorescence , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/ultrastructure , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/ultrastructure , Polymerase Chain Reaction , Time Factors , Tumor Burden
12.
Cancer Res ; 69(2): 422-30, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19147554

ABSTRACT

Primary cilia have been proposed to participate in the modulation of growth factor signaling pathways. In this study, we determined that ciliogenesis is suppressed in both pancreatic cancer cells and pancreatic intraepithelial neoplasia (PanIN) lesions in human pancreatic ductal adenocarcinoma (PDAC). Primary cilia were absent in these cells even when not actively proliferating. Cilia were also absent from mouse PanIN cells in three different mouse models of PDAC driven by an endogenous oncogenic Kras allele. Inhibition of Kras effector pathways restored ciliogenesis in a mouse pancreatic cancer cell line, raising the possibility that ciliogenesis may be actively repressed by oncogenic Kras. By contrast, normal duct, islet, and centroacinar cells retained primary cilia in both human and mouse pancreata. Thus, arrested ciliogenesis is a cardinal feature of PDAC and its precursor PanIN lesions, does not require ongoing proliferation, and could potentially be targeted pharmacologically.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/pathology , Precancerous Conditions/pathology , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/ultrastructure , Cell Growth Processes/physiology , Cell Line, Tumor , Centrosome/pathology , Cilia/pathology , Epithelial Cells/pathology , Genes, ras , Humans , Mice , Mice, Transgenic , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/ultrastructure , Precancerous Conditions/genetics , Precancerous Conditions/ultrastructure , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism
13.
Dig Dis Sci ; 50(8): 1402-9, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16110828

ABSTRACT

In our previous study, dissociation factor (DF) and mitogen-activated protein kinase kinase 2 (MEK2) were isolated as factors relating to cancer cell dissociation in pancreatic cancer cells. On the other hand, tight junction protein zonula occludens 1 (ZO-1) has been indicated to be involved in carcinogenesis. In this study, the expression of ZO-1 and a downstream kinase of MEK2, extracellular signal-regulated kinase 2 (ERK2), was analyzed to clarify the regulatory mechanism of cell dissociation in pancreatic cancer cells. Two hamster (PC-1.0 and PC-1) and two human (AsPC-1 and CAPAN-2) pancreatic cancer cell lines were used. Immunocytochemical study was performed using anti-ZO-1, ERK2, and phosphorylated ERK1/2 (p-ERK1/2) antibodies. DF treatment obviously disrupted ZO-1 expression at the sites of cell-cell contact and markedly induced ERK2 and p-ERK1/2 expression, as well as the dissociation of cell clones in PC-1 and CAPAN-2 cells. In contrast, U0126 (a MEK1/2 inhibitor) treatment significantly induced the peripheral distribution of ZO-1 as well as cell aggregation in PC-1.0 and AsPC-1 cells, which usually grew as single cells, but seriously suppressed ERK2 and p-ERK1/2 expression. We conclude that redistribution of ZO-1 is closely correlated with cell dissociation status in pancreatic cancer cells through activation of ERK2.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Pancreatic Neoplasms/metabolism , Phosphoproteins/metabolism , Signal Transduction/physiology , Animals , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/ultrastructure , Cell Adhesion/physiology , Cell Line, Tumor/metabolism , Cricetinae , Humans , Neoplasm Invasiveness , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/ultrastructure , Zonula Occludens-1 Protein
14.
J Korean Med Sci ; 20(3): 516-20, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15953882

ABSTRACT

Osteoclast-like giant cell tumor of the pancreas is a very rare neoplasm, of which the histiogenesis remains controversial. A 63-yr-old woman was hospitalized for evaluation of epigastric pain. An abdominal computerized tomography revealed the presence of a large cystic mass, arising from the tail of pancreas. A distal pancreatectomy with splenectomy was performed. Histologically, the tumor was composed of mononuclear stromal cells intermingled with osteclast-like giant cells. In addition, there was a small area of moderately to well differentiated ductal adenocarcinoma. The final pathologic diagnosis was osteoclast-like giant cell tumor of the pancreas with ductal adenocarcinoma. Here, we describe the histopathological, immunohistochemical, ultrastructural and molecular biological findings of this tumor with review of the literature pertaining to this condition.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Giant Cell Tumors/pathology , Osteoclasts/pathology , Pancreatic Neoplasms/pathology , Antigens, CD/analysis , Antigens, Differentiation, Myelomonocytic/analysis , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/ultrastructure , Diagnosis, Differential , Female , Giant Cell Tumors/metabolism , Giant Cell Tumors/ultrastructure , Humans , Immunohistochemistry , Keratins/analysis , Microscopy, Electron , Middle Aged , Mucin-1/analysis , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/ultrastructure , Proliferating Cell Nuclear Antigen/analysis , Vimentin/analysis
15.
J Mol Histol ; 35(4): 355-62, 2004 May.
Article in English | MEDLINE | ID: mdl-15503809

ABSTRACT

When grown on permeable supports, pancreatic duct adenocarcinoma CAPAN-1 cells establish very high values of transepithelial resistance (TER). The addition of ethanol produced a dose-related, reversible drop in the TER of these cells, ranging from 15% (with 1% ethanol) to 65% (with 10% ethanol). The ethanol effect was rapid and reversible. The resistance decrease was associated with an increase in monolayer permeability to mannitol. No significant decrease in cell ATP was detected for ethanol concentrations lower than 7%. Confocal vertical sections of calcein-loaded monolayers of CAPAN-1 cells, grown on plasticware, showed a progressive deflation of domes detectable after 5 min of treatment with 2% ethanol. Incubation in an ethanol-free medium caused a progressive dome restoration. Immunocytochemical analysis of ethanol-treated cells indicated that ZO-1 and occludin exhibited clear cut distribution changes while the perijunctional actin pattern was slightly modified. Electron microscopy showed that a discrete intercellular space was detectable between adjacent ethanol-treated cells but not between control cells. These data indicate that ethanol is a tight junction barrier opener in pancreatic duct cells.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Ethanol/pharmacology , Pancreatic Neoplasms/metabolism , Solvents/pharmacology , Tight Junctions/metabolism , Actins/metabolism , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/ultrastructure , Cell Line, Tumor , Humans , Immunohistochemistry , Mannitol/metabolism , Membrane Proteins/metabolism , Microscopy, Electron, Transmission , Occludin , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/ultrastructure , Permeability/drug effects , Phosphoproteins/metabolism , Tight Junctions/drug effects , Zonula Occludens-1 Protein
16.
J Submicrosc Cytol Pathol ; 35(4): 343-9, 2003 Oct.
Article in English | MEDLINE | ID: mdl-15137676

ABSTRACT

Pancreatic undifferentiated carcinomas with a neoplastic mesenchymal component (carcinosarcoma) have not been well described to date. The author experienced an autopsy case of a unique pancreatic ductal adenocarcinoma with carcinosarcomatous histology. The patient was a 90 year old Japanese male who died of cahexia with generalized tumor extension. Post-mortem examinations revealed some distinctive or representative components discerned in the tumor tissue. One was the well differentiated ductal adenocarcinoma. The second and the major finding was undifferentiated short spindle shaped or small round sarcomatous cells, which lacked an epithelial nature but showed positivity for CD10+, CD56+, Ki67++, p53++, and were focally positive for Desmin and vimentin. These two components were mixed and constituted the histology of the carcinosarcoma. In another area, anaplastic, large, pleomorphic tumor cells showed the focal immunohistochemical distribution of alpha-feto-protein and human chorionic gonadotropin. An ultrastructural study revealed adenocarcinoma cells with apical mucin secreting granules and well developed ductal differentiation, whereas undifferentiated sarcomatous cells showed primitive fibroblastic or mesenchymal characters without specific differentiation. Conclusively these findings suggested that this well differentiated adenocarcinoma gradually enlarged, accumulated genetic alternations, and then transformed into large and undifferentiated tumor cells, rapidly growing small sarcomatous cells, and a histology of carcinosarcoma.


Subject(s)
Adenocarcinoma/pathology , Carcinoma, Pancreatic Ductal/pathology , alpha-Fetoproteins/metabolism , Adenocarcinoma/ultrastructure , Aged , Aged, 80 and over , Carcinoma, Pancreatic Ductal/ultrastructure , Chorionic Gonadotropin, beta Subunit, Human , Humans , Immunohistochemistry , Male , Microscopy, Electron
17.
Cancer Detect Prev ; 24(6): 572-8, 2000.
Article in English | MEDLINE | ID: mdl-11198271

ABSTRACT

Differentiation of benign from malignant pancreatic endocrine tumors by existing clinical, biochemical, histologic, and cytologic criteria is difficult. We immunohistochemically localized pancreatic secretory trypsin inhibitor (PSTI) in 28 pancreatic endocrine tumors (13 benign, 15 malignant). PSTI-immunoreactive cells were detected in nine endocrine tumors. Immunoreactivity in these tumors was detected in nearly all tumor cells in five cases, scattered cells in two cases, and a few cells in two cases. All positive cases were malignant, and eight were equal to or larger than 10 cm. Serum concentrations of PSTI were markedly elevated in the two patients so tested. PSTI may be a specific immunohistochemical marker for malignant pancreatic endocrine tumors.


Subject(s)
Carcinoma, Pancreatic Ductal/chemistry , Glucagonoma/chemistry , Insulinoma/chemistry , Neoplasm Proteins/analysis , Pancreatic Neoplasms/chemistry , Trypsin Inhibitor, Kazal Pancreatic/analysis , Adult , Amylases/analysis , Biomarkers, Tumor/analysis , Carcinoma, Pancreatic Ductal/classification , Carcinoma, Pancreatic Ductal/ultrastructure , Chromogranin A , Chromogranins/analysis , DNA, Neoplasm/analysis , Glucagonoma/ultrastructure , Humans , Immunoenzyme Techniques , Insulinoma/ultrastructure , Ki-67 Antigen/analysis , Neoplasm Metastasis , Pancreatic Elastase/analysis , Pancreatic Hormones/analysis , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/ultrastructure , Pancreatic Polypeptide/analysis , Paraffin Embedding , Serotonin/analysis , Synaptophysin/analysis , Trypsin/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...