Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
2.
Int J Cancer ; 134(11): 2572-82, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24214898

ABSTRACT

Hypoxia and dysfunctional tumor vessels represent a prominent feature of pancreatic cancer, being, at least in part, responsible for chemotherapy resistance and immune suppression in these tumors. We tested whether the increase of oxygen delivery induced in vivo by myo-inositol trispyrophosphate (ITPP) can reverse hypoxia, control tumor growth and improve chemotherapy response. Tumor size, metastatic development (microcomputed tomography scan follow-up) and the survival of rats and nude or NOD.SCID mice, (bearing syngenic rat and MiaPaCa2- or patient-derived pancreatic tumors), were determined on ITPP and/or gemcitabine treatment. Partial oxygen pressure, expression of angiogenic factors and tumor histology were evaluated. Infiltration and oxidative status of immune cells, as well as chemotherapy penetration in tumors, were determined by fluorescence-activated cell sorting, fluorometry, nitric oxide release assays, Western blot and confocal microscopy. Weekly intravenous ITPP application resulted in the inhibition of metastasis development and restricted primary tumor growth, showing a superior effect on the rats' survival compared with gemcitabine. ITPP treatment restored tumor normoxia and caused a reduction in hypoxia inducible factor-1α levels, with subsequent VEGF and Lox downregulation, resulting in improved vessel structure and decreased desmoplasia. The latter effects translated into elevated immune cells influx and improved susceptibility to gemcitabine treatment. Growth of human pancreatic tumor xenografts was strongly inhibited by administration of ITPP. ITPP exploits a two-stage mechanism causing rapid, early and sustainable late stage normoxia. This is due to the angiogenic factor modulation and vascular normalization, leading to enhanced chemotherapy delivery and synergistic life prolongation, on combination with low doses of gemcitabine.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Deoxycytidine/analogs & derivatives , Hypoxia/drug therapy , Inositol Phosphates/therapeutic use , Liver Neoplasms/drug therapy , Pancreatic Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Blotting, Western , Cell Proliferation/drug effects , Deoxycytidine/therapeutic use , Drug Synergism , Fluorescent Antibody Technique , Humans , Hypoxia/metabolism , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neovascularization, Pathologic , Oxygen/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , RNA, Messenger/genetics , Rats , Rats, Inbred Lew , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Gemcitabine
3.
Oncogene ; 33(26): 3401-10, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-23912455

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is usually incurable. Contrary to genetic mechanisms involved in PDAC pathogenesis, epigenetic alterations are ill defined. Here, we determine the contribution of epigenetically silenced genes to the development of PDAC. We analyzed enriched, highly methylated DNAs from PDACs, chronic pancreatitis (CP) and normal tissues using CpG island microarrays and identified WNK2 as a prominent candidate tumor suppressor gene being downregulated early in PDAC development. WNK2 was further investigated in tissue microarrays, methylation analysis of early pancreatic intraepithelial neoplasia (PanIN), mouse models for PDAC and pancreatitis, re-expression studies after demethylation, and cell growth assays using WNK2 overexpression. Demethylation assays confirmed the link between methylation and expression. WNK2 hypermethylation was higher in tumor than in surrounding inflamed tissues and was observed in PanIN lesions as well as in a PDAC mouse model. WNK2 mRNA and protein expressions were lower in PDAC and CP compared with normal tissues both in patients and mouse models. Overexpression of WNK2 led to reduced cell growth, and WNK2 expression in tissues correlated negatively with pERK1/2 expression, a downstream target of WNK2 responsible for cell proliferation. Downregulation of WNK2 by promoter hypermethylation occurs early in PDAC pathogenesis and may support tumor cell growth via the ERK-MAPK pathway.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , DNA Methylation/genetics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms/genetics , Protein Serine-Threonine Kinases/genetics , Animals , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation , CpG Islands/genetics , Down-Regulation , Extracellular Signal-Regulated MAP Kinases/biosynthesis , Female , Humans , Male , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Pancreatic Neoplasms/pathology , Pancreatitis, Chronic/genetics , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/biosynthesis , RNA, Messenger/biosynthesis
4.
Cell Death Dis ; 4: e627, 2013 May 09.
Article in English | MEDLINE | ID: mdl-23661005

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) is one of the most lethal malignancies. Cancer stem cells (CSCs), which are not targeted by current therapies, may be the reason for pronounced therapy resistance. A new treatment option in phase II trials is cabozantinib that inhibits the pancreatic CSC surface marker and tyrosine kinase receptor c-Met. The purpose of this study was to evaluate the effect of cabozantinib to stem-like features and therapy resistance. Established PDA cell lines, a gemcitabine-resistant subclone, non-malignant pancreatic ductal cells and primary spheroidal cultures from patient tumors were analyzed by MTT-assay, flow cytometry, colony and spheroid formation assays, western blotting, qRT-PCR, antibody protein array, immunohistochemistry and morphological features. Cabozantinib inhibited viability and spheroid formation and induced apoptosis in malignant cells with minor effects in non-malignant cells. After long-term cabozantinib treatment, PDA cells had altered anti- and pro-apoptotic signaling, but still responded to cabozantinib, as apoptosis only slightly decreased and viability only slightly increased suggesting a low resistance-inducing potential of cabozantinib. In parallel, c-Met expression and the pluripotency transcription factor SOX2 were downregulated, which might counteract development of full therapy resistance in long-term treated subclones. In single-treatment studies, cabozantinib increased efficacy of gemcitabine. Most importantly, cabozantinib strongly induced apoptosis and reduced viability in PDA cell lines, which are completely resistant toward gemcitabine. In primary, CSC-enriched spheroidal cultures cabozantinib downregulated CSC markers SOX2, c-Met and CD133 and induced apoptosis. These findings suggest that the clinical use of cabozantinib may be more effective than current chemotherapeutics.


Subject(s)
Anilides/pharmacology , Apoptosis/drug effects , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Pyridines/pharmacology , Stem Cells/metabolism , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Down-Regulation , Drug Resistance, Neoplasm/drug effects , Humans , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-met/metabolism , SOXB1 Transcription Factors/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/drug effects , Gemcitabine
5.
Eur Surg Res ; 48(3): 131-8, 2012.
Article in English | MEDLINE | ID: mdl-22572771

ABSTRACT

BACKGROUND/AIMS: We aimed to analyze substance P (SP) and neprilysin (NEP), the membrane metallopeptidase that degrades SP, in chronic pancreatitis (CP). METHODS: SP and NEP mRNA levels were analyzed by qRT-PCR in tissue samples from 30 patients with CP and 8 organ donors. In addition, SP serum levels were determined before and after surgery in the same patients, by means of a competitive ELISA assay. Genetic and epigenetic analyses of the NEP gene were also performed. RESULTS: SP mRNA expression levels were higher in CP tissues compared to controls (p = 0.0152), while NEP mRNA showed no significant differences between CP and healthy subjects (p = 0.2102). In CP patients, SP serum levels correlated with those in tissue, and after surgical resection SP serum levels were reduced compared to the preoperative values. Failure of NEP to overexpress in CP tissues was associated with significant miR-128a overexpression (p = 0.02), rather than with mutations in the NEP coding region or the presence of hypermethylation sites in the NEP promoter region. CONCLUSION: Tissue and serum levels of SP were increased in CP, while NEP levels remained unaltered. In an SP/NEP-mediated pathway, it would appear that NEP fails to provide adequate surveillance of SP levels. Failure of NEP to overexpress could be associated with miRNA regulation.


Subject(s)
Neprilysin/physiology , Pancreatitis, Chronic/etiology , Substance P/physiology , Adult , Aged , DNA Methylation , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged , Neprilysin/blood , Neprilysin/genetics , Pancreatitis, Chronic/blood , Promoter Regions, Genetic , RNA, Messenger/analysis , Substance P/blood , Substance P/genetics
6.
Br J Cancer ; 105(2): 288-95, 2011 Jul 12.
Article in English | MEDLINE | ID: mdl-21712827

ABSTRACT

BACKGROUND: The transcription factor HOXC8 regulates many genes involved in tumour progression. This study was to investigate the role of HOXC8 in pancreatic ductal adenocarcinoma (PDAC) growth and metastasis. METHODS: The Hoxc8 expression was determined in 15 PDAC cell lines and human specimens by RT-polymerase chain reaction and/or immunohistochemistry. The effects of HOXC8 silencing by RNA interference were investigated by functional tests. RESULTS: The Hoxc8 mRNA expression in PDAC cell lines was negatively related to their growth in vivo. Except for Suit2-007 cells, only those with low Hoxc8 mRNA expression grew in nude rats. Successful down-regulation of HOXC8 expression caused increased proliferation, migration (P ≤ 0.05) and colony formation (P ≤ 0.05) in Suit2-007, Panc-1 and MIA PaCa-2 PDAC cells, respectively. The Hoxc8 mRNA levels in diseased human pancreas tissues were significantly increased over normal in PDAC and autoimmune chronic pancreatitis specimens (P<0.01, respectively), but negatively related to tumour stage (P=0.09). In primary and metastatic tumour samples, immunohistochemical staining for HOXC8 was stronger in surrounding than in neoplastic tissues. Furthermore, grading of primary carcinomas was negatively associated with HOXC8 staining (P=0.03). Liver metastases showed the lowest HOXC8 expression of all neoplastic lesions. CONCLUSION: These data indicate that HOXC8 expression is inversely related to PDAC progression and metastasis.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Homeodomain Proteins/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Case-Control Studies , Cell Line, Tumor , Disease Progression , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/physiology , Humans , Male , Middle Aged , Neoplasm Metastasis , RNA, Small Interfering/pharmacology , Rats , Rats, Nude , Xenograft Model Antitumor Assays , Young Adult
7.
Br J Cancer ; 103(10): 1571-9, 2010 Nov 09.
Article in English | MEDLINE | ID: mdl-20978507

ABSTRACT

PURPOSE: Tumour hypoxia activates hypoxia-inducible factor-1 (HIF-1) and indluences angiogenesis, cell survival and invasion. Prolyl hydroxylase-3 (PHD3) regulates degradation of HIF-1α. The effects of PHD3 in tumour growth are largely unknown. EXPERIMENTAL DESIGN: PHD3 expression was analysed in human pancreatic cancer tissues and cancer cell lines by real-time quantitative PCR and immunohistochemistry. PHD3 overexpression was established by stable transfection and downregulation by short interfering RNA technology. VEGF was quantified by enzyme-linked immunosorbent assay. Matrigel invasion assays were performed to examine tumour cell invasion. Apoptosis was measured by annexin-V staining and caspase-3 assays. The effect of PHD3 on tumour growth in vivo was evaluated in an established orthotopic murine model. RESULTS: PHD3 was upregulated in well-differentiated human tumours and cell lines, and regulated hypoxic VEGF secretion. PHD3 overexpression mediated tumour cell growth and invasion by induction of apoptosis in a nerve growth factor-dependent manner by the activation of caspase-3 and phosphorylation of focal adhesion kinase HIF-1 independently. In vivo, PHD3 inhibited tumour growth by abrogation of tumour angiogenesis. CONCLUSION: Our results indicate essential functions of PHD3 in tumour growth, apoptosis and angiogenesis and through HIF-1-dependent and HIF-1-independent pathways.


Subject(s)
Dioxygenases/genetics , Neovascularization, Pathologic/pathology , Pancreatic Neoplasms/pathology , Adenocarcinoma/pathology , Adenocarcinoma/surgery , Animals , Annexin A5/analysis , Apoptosis , Carcinoma, Pancreatic Ductal/enzymology , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/surgery , Caspase 3/metabolism , Cell Differentiation , Cell Line, Tumor , Dioxygenases/physiology , Enzyme-Linked Immunosorbent Assay , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Hypoxia-Inducible Factor-Proline Dioxygenases , Mice , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/surgery , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous , Up-Regulation , Vascular Endothelial Growth Factor A/analysis
8.
Cancer Immunol Immunother ; 59(9): 1389-400, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20514540

ABSTRACT

Despite spontaneous or vaccination-induced immune responses, pancreatic cancer remains one of the most deadly immunotherapy-resistant malignancies. We sought to comprehend the spectrum of pancreatic tumor-associated antigens (pTAAs) and to assess the clinical relevance of their immunogenicity. An autologous SEREX-based screening of a cDNA library constructed from a pancreatic T3N0M0/GIII specimen belonging to a long-term survivor (36 months) revealed 18 immunogenic pTAA. RT-PCR analysis displayed broad distribution of the identified antigens among normal human tissues. PNLIPRP2 and MIA demonstrated the most distinct pancreatic cancer-specific patterns. ELISA-based screening of sera for corresponding autoantibodies revealed that although significantly increased, the immunogenicity of these molecules was not a common feature in pancreatic cancer. QRT-PCR and immunohistochemistry characterized PNLIPRP2 as a robust acinar cell-specific marker whose decreased expression mirrored the disappearance of parenchyma in the diseased organ, but was not related to the presence of PNLIPRP2 autoantibodies. Analyses of MIA-known to be preferentially expressed in malignant cells-surprisingly revealed an inverse correlation between intratumoral gene expression and the emergence of autoantibodies. MIA(high) patients were autoantibody-negative and had shorter median survival when compared with autoantibody-positive MIA(low) patients (12 vs. 34 months). The observed pTAA spectrum comprised molecules associated with acinar, stromal and malignant structures, thus presenting novel targets for tumor cell-specific therapies as well as for approaches based on the bystander effects. Applying the concept of cancer immunoediting to interpret relationships between gene expression, antitumor immune responses, and clinical outcome might better discriminate between past and ongoing immune responses, consequently enabling prognostic stratification of patients and individual adjustment of immunotherapy.


Subject(s)
Adenocarcinoma/diagnosis , Adenocarcinoma/immunology , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/immunology , Adenocarcinoma/blood , Adenocarcinoma/genetics , Adenocarcinoma/mortality , Aged , Antigens, Tumor-Associated, Carbohydrate/genetics , Antigens, Tumor-Associated, Carbohydrate/immunology , Antigens, Tumor-Associated, Carbohydrate/metabolism , Autoantibodies/blood , Cloning, Molecular , Enzyme-Linked Immunosorbent Assay , Female , Gene Library , Humans , Immunochemistry , Lipase/genetics , Lipase/immunology , Lipase/metabolism , Male , Middle Aged , Neoplasm Staging , Pancreas/metabolism , Pancreas/pathology , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/mortality , Prognosis
9.
Br J Cancer ; 99(7): 1064-73, 2008 Oct 07.
Article in English | MEDLINE | ID: mdl-18781173

ABSTRACT

Pancreatic cancer has an abysmal prognosis. Targets for early detection, prevention and therapy are desperately needed. Inflammatory pathways have an important impact on tumour growth and progression. Expression of BLT2 (the second leukotriene B(4) receptor) was investigated by real-time RT-PCR and immunohistochemistry. Cell proliferation was studied after stable transfection with BLT2, after treatment with siRNA and Compound A as an agonist. BLT2 is expressed in all pancreatic cancer cell lines. Results from real-time RT-PCR revealed significant overexpression of BLT2 in malignant intraductal papillary mucinous neoplasias (IPMNs) and pancreatic adenocarcinoma. Intense staining was evident in IPMNs, infiltrating tumour cells and advanced pancreatic intraepithelial neoplasias (PanINs) but not in normal ductal cells. Overexpression of BLT2 as well as stimulation of Colo357, Panc-1 and AsPC1 cells with Compound A caused a significant increase in tumour cell proliferation, an effect reversed after siRNA treatment. This study demonstrates for the first time the expression of BLT2 in the pancreas and overexpression in pancreatic cancers and malignant IPMNs in particular. Upregulation of BLT2 is already evident in precursor lesions (PanINs, IPMNs). Overexpression of this receptor leads to significant growth stimulation. Therefore, we suggest BLT2 as a new target for chemoprevention and therapy for pancreatic cancer.


Subject(s)
Cell Proliferation , Pancreatic Neoplasms/metabolism , Receptors, Leukotriene B4/metabolism , Base Sequence , Cell Line, Tumor , DNA Primers , Humans , Immunohistochemistry , Leukotriene B4/metabolism , Ligands , Pancreatic Neoplasms/pathology , Pancreatitis/metabolism , RNA, Small Interfering , Receptors, Leukotriene B4/genetics , Reverse Transcriptase Polymerase Chain Reaction
10.
Clin Exp Immunol ; 154(2): 216-23, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18778363

ABSTRACT

The chemokine receptor CXCR6 has been described on lymphoid cells and is thought to participate in the homing of activated T-cells to non-lymphoid tissue. We now provide evidence that the chemokine receptor CXCR6 is also expressed by activated polymorphonuclear neutrophils (PMN) in vivo: Examination of biopsies derived from patients with pancreatic carcinoma by confocal laser scan microscopy revealed a massive infiltration of PMN that expressed CXCR6, while PMN of the peripheral blood of these patients did not. To answer the question whether CXCR6 expression is a property of infiltrated and activated PMN, leucocytes were collected from patients with localized soft tissue infections in the course of the wound debridement. By cytofluorometry, the majority of these cells were identified as PMN. Up to 50% of these PMN were also positive for CXCR6. Again, PMN from the peripheral blood of these patients were nearly negative for CXCR6, as were PMN of healthy donors. In a series of in vitro experiments, up-regulation of CXCR6 on PMN of healthy donors by a variety of cytokines was tested. So far, a minor, although reproducible, effect of tumour necrosis factor (TNFalpha) was seen: brief exposure with low-dose TNFalpha induced expression of CXCR6 on the surface of PMN. Furthermore, we could show an increased migration of PMN induced by the axis CXCL16 and CXCR6. In summary, our data provide evidence that CXCR6 is not constitutively expressed on PMN, but is up-regulated under inflammatory conditions and mediates migration of CXCR6-positive PMN.


Subject(s)
Bacterial Infections/immunology , Neoplasm Proteins/metabolism , Neutrophils/immunology , Pancreatic Neoplasms/immunology , Receptors, Chemokine/metabolism , Receptors, Virus/metabolism , Acute Disease , Chemotaxis, Leukocyte/immunology , Humans , Ligands , Microscopy, Confocal/methods , Neutrophil Activation/immunology , Osteomyelitis/immunology , Prosthesis-Related Infections/immunology , Receptors, CXCR6 , Soft Tissue Infections/immunology , Tumor Cells, Cultured , Up-Regulation/immunology
11.
Br J Cancer ; 99(5): 760-7, 2008 Sep 02.
Article in English | MEDLINE | ID: mdl-18728667

ABSTRACT

Efficacy of chemotherapy for pancreatic cancer may be improved by tailoring it to individual chemosensitivity profiles. Identification of nonresponders before initiation of treatment may help to avoid side effects. In this study, primary pancreatic cancer cells were isolated from 18 patients undergoing pancreaticoduodenectomy for pancreatic cancer. Eight commonly used pancreatic cancer cell lines were used as controls. Ex vivo chemosensitivity for gemcitabine, 5-fluorouracil, mitomycin-C, cisplatinum, oxaliplatinum, paclitaxel and a combination of gemcitabine with oxaliplatinum or mitomycin-C was determined using a cellular ATP-based tumour chemosensitivity assay (ATP-TCA). Quantitative real-time-polymerase chain reaction was performed to determine RNA expression levels of genes implicated in chemoresistance. Chemosensitivity towards cytotoxic agents was highly variable in primary pancreatic cancer cells and pancreatic cancer cell lines. ATP-TCA results for gemcitabine correlated to the tissue expression of human equilibrative nucleoside transporter-1 (hENT1). Time to relapse in patients with gemcitabine-sensitive tumours was significantly higher than in patients with chemoresistant pancreatic cancers (P=0.01; 71 vs 269 days). Furthermore, time to relapse in gemcitabine-treated patients was related to hENT1 expression (P=0.0067). Thus, chemosensitivity testing using ATP-TCA in pancreatic cancer is feasible and correlated with time to relapse in gemcitabine-treated patients. This suggests that ATP-TCA testing could be used as a decision-making tool in the adjuvant treatment of pancreatic cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Deoxycytidine/analogs & derivatives , Gene Expression Profiling , Pancreatic Neoplasms/drug therapy , Adult , Aged , Cell Line, Tumor , Deoxycytidine/therapeutic use , Drug Screening Assays, Antitumor , Female , Humans , Male , Middle Aged , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Polymerase Chain Reaction , Gemcitabine
12.
Br J Cancer ; 99(4): 622-31, 2008 Aug 19.
Article in English | MEDLINE | ID: mdl-18665180

ABSTRACT

Little is known about the factors that enable the mobilisation of human mesenchymal stem cells (MSC) from the bone marrow into the blood stream and their recruitment to and retention in the tumour. We found specific migration of MSC towards growth factors present in pancreatic tumours, such as PDGF, EGF, VEGF and specific inhibitors Glivec, Erbitux and Avastin interfered with migration. Within a few hours, MSC migrated into spheroids consisting of pancreatic cancer cells, fibroblasts and endothelial cells as measured by time-lapse microscopy. Supernatant from subconfluent MSC increased sprouting of HUVEC due to VEGF production by MSC itself as demonstrated by RT-PCR and ELISA. Only few MSCs were differentiated into endothelial cells in vitro, whereas in vivo differentiation was not observed. Lentiviral GFP-marked MSCs, injected in nude mice xenografted with orthotopic pancreatic tumours, preferentially migrated into the tumours as observed by FACS analysis of green fluorescent cells. By immunofluorescence and intravital microscopic studies, we found the interaction of MSC with the endothelium of blood vessels. Mesenchymal stem cells supported tumour angiogenesis in vivo, that is CD31(+) vessel density was increased after the transfer of MSC compared with siVEGF-MSC. Our data demonstrate the migration of MSC toward tumour vessels and suggest a supportive role in angiogenesis.


Subject(s)
Mesenchymal Stem Cells/metabolism , Neovascularization, Pathologic/metabolism , Pancreatic Neoplasms/blood supply , Vascular Endothelial Growth Factor A/metabolism , Actins/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacology , Benzamides , Bevacizumab , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation , Cells, Cultured , Cetuximab , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Epidermal Growth Factor/antagonists & inhibitors , Epidermal Growth Factor/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Imatinib Mesylate , Lentivirus/genetics , Male , Mesenchymal Stem Cell Transplantation , Mice , Mice, Nude , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Piperazines/pharmacology , Platelet-Derived Growth Factor/antagonists & inhibitors , Platelet-Derived Growth Factor/metabolism , Pyrimidines/pharmacology , Spheroids, Cellular/pathology , Transplantation, Heterologous , Umbilical Veins/cytology , Umbilical Veins/drug effects , Umbilical Veins/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors
13.
J Pathol ; 215(2): 195-203, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18421760

ABSTRACT

In chronic pancreatitis (CP), both the progressive loss of acinar parenchyma and aggressive fibro-inflammatory reactions ultimately lead to irreversible organ destruction. Dying cells are normally removed by macrophages and elimination is associated with anti-inflammatory cytokine switch. We investigated whether defective clearance of damaged acini by macrophages such as compromised phagocytosis or altered cytokine reaction occurs in CP and thus represents a causative link between acinar loss and fibro-inflammation. In a checkerboard-like co-culture system, we assessed normal and CP macrophages for their phagocytic and cytokine responses to dying pancreatic acinar cells of normal or CP origin by FACS, confocal microscopy, QRT-PCR, and ELISA. In CP, phagocytosis of apoptotic acini by macrophages was not impaired; however, the associated cytokine responses were gradually perturbed. Most interestingly, only normal acini suppressed TGFbeta1 expression and accumulation specifically in normal macrophage cultures, while CP acini lost this ability. Both types of apoptotic acini induced pro-inflammatory cytokine bursts of varying strength in both types of macrophages; however, the most significant difference (more than 50-fold higher expression of IL-1beta, IL-6, and IL-8) was evident between CP/CP and normal/normal combinations, indicating that acinar and macrophage alterations synergistically lead to the ultimate CP-specific bias. In combination with in situ data comparing circulating inflammatory cells to pancreatic resident ones, our results indicate that cytokine expression in inflammatory cells undergoes spatiotemporal modulation, most likely through a successive interplay of acinar, stromal, and circulating factors. Thus, clearance of injured pancreatic acini by macrophages is associated with a unique cytokine reaction which may constitute a basis for progression of SAPE (sentinel acute pancreatitis event) to the irreversible fibro-inflammation in CP.


Subject(s)
Macrophages/physiology , Pancreas/immunology , Pancreatitis, Chronic/immunology , Apoptosis , Coculture Techniques , Enzyme-Linked Immunosorbent Assay/methods , Flow Cytometry , Humans , Interleukin-1/immunology , Interleukin-6/immunology , Interleukin-8/immunology , Macrophages/immunology , Microscopy, Confocal , Pancreas/pathology , Pancreatitis, Chronic/pathology , Phagocytosis , Polymerase Chain Reaction/methods , Tumor Necrosis Factor-alpha/immunology
14.
J Cell Mol Med ; 11(5): 1162-74, 2007.
Article in English | MEDLINE | ID: mdl-17979891

ABSTRACT

ADAM8 belongs to a family of transmembrane proteins implicated in cell-cell interactions, proteolysis of membrane proteins, and various aspects of carcinogenesis. In the present study, we aimed to evaluate the expression and function of ADAM8 in pancreatic cancer. ADAM8 mRNA levels were analysed by quantitative RT-PCR and correlated to patient survival. Immunohistochemistry was performed to localize ADAM8 in pancreatic tis-sues. Silencing of ADAM8 expression was carried out by transfection with specific siRNA oligonucleotides. Cell growth and invasion assays were used to assess the functional consequences of ADAM8 silencing. SELDI-TOF-MS was performed to detect the proteolytic activity of ADAM8 in pancreatic cancer cells. ADAM8 mRNA was significantly overexpressed in pancreatic ductal adenocarcinoma (PDAC) compared with normal pancreatic tissues (5.3-fold increase; P= 0.0008), and high ADAM8 mRNA and protein expression levels correlated with reduced survival time of PDAC patients (P= 0.048 and P= 0.065, respectively). Silencing of ADAM8 expression did not significantly influence pancreatic cancer cell growth but suppressed invasiveness. In addition, decreased proteolytic activity was measured in cell culture supernatants following silencing of ADAM8. In conclusion, ADAM8 is overexpressed in PDAC, influences cancer cell invasiveness and correlates with reduced survival, suggesting that ADAM8 might be a potential target in pancreatic cancer therapy.


Subject(s)
ADAM Proteins/genetics , Carcinoma, Pancreatic Ductal/pathology , Membrane Proteins/genetics , Pancreatic Neoplasms/pathology , ADAM Proteins/chemistry , ADAM Proteins/metabolism , Adult , Carcinoma, Pancreatic Ductal/genetics , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Middle Aged , Neoplasm Invasiveness , Pancreatic Neoplasms/genetics , Protein Processing, Post-Translational , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Survival Analysis
15.
J Pathol ; 208(5): 673-85, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16450333

ABSTRACT

Tenascin C (TNC) is a component of the provisional extracellular matrix (ECM) that characterizes solid tumours. Cell surface annexin II is a high-affinity receptor for large TNC splice variants. The aim of this study was to analyse whether TNC and annexin II play a role in the development of pancreatic ductal adenocarcinoma (PDAC). PDAC is characterized by a rich ECM populated by pancreatic stellate cells, which play a crucial role in pancreatic desmoplasia. The mRNA and protein levels of TNC and of annexin II were analysed in pancreatic tissues by DNA array, quantitative reverse transcriptase-polymerase chain reaction (QRT-PCR) and immunohistochemistry. TNC large splice variants were detected by RT-PCR. Enzyme linked immunosorbent assay (ELISA) was used to measure TNC levels in serum and culture supernatants. TNC and annexin II mRNA levels were significantly higher in pancreatic cancer tissues than in the normal pancreas. TNC expression was detected with increased frequency in the progression from PanIN-1 lesions to PDAC, and a parallel switch from cytoplasmic to cell surface expression of annexin II was observed. Large TNC transcripts were found in pancreatic cancer and in chronic pancreatitis, but not in the normal pancreas. TNC expression was demonstrated in pancreatic stellate cells, where it could be induced by tumour necrosis factor alpha (TNFalpha), transforming growth factor beta1 (TGF-beta1) and by cancer cell supernatants supplemented with TGF-beta1. In conclusion, the expression of TNC and cell surface annexin II increases in the progression from low-grade PanIN lesions to pancreatic cancer. Pancreatic stellate cells are identified as a source of TNC in pancreatic tissues, possibly under the influence of soluble factors released by the tumour cells.


Subject(s)
Adenocarcinoma/metabolism , Annexin A2/metabolism , Cell Transformation, Neoplastic/metabolism , Pancreatic Neoplasms/metabolism , Tenascin/metabolism , Adenocarcinoma/genetics , Annexin A2/genetics , Blotting, Western , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunoenzyme Techniques , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/genetics , Pancreatitis, Chronic/metabolism , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Tenascin/genetics , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta1 , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology
16.
J Clin Pathol ; 59(1): 21-7, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16394277

ABSTRACT

BACKGROUND: Recurrent inflammation in chronic pancreatitis (CP) is not well understood. AIMS: To investigate whether decorin, an extracellular matrix (ECM) proteoglycan with macrophage modulating activity, is a pathogenic factor allowing diseased pancreatic stroma to sustain inflammation by affecting the cytokine profile of accumulating inflammatory cells. METHODS: Decorin was examined in 18 donors and 32 patients with CP by quantitative reverse transcription polymerase chain reaction (QRT-PCR), western blotting, and immunohistochemistry of pancreatic specimens. QRT-PCR was used to assess cytokine expression in donor peripheral blood mononuclear cells (PBMC), exposed or not to decorin in vitro, and to compare it with the cytokine profile of circulating and resident mononuclear cells (MNC) of patients with CP. RESULTS: In CP, desmoplasia is associated with overexpression of decorin in the growing ECM and enlarged pancreatic nerves. In culture, exposure of MNC to decorin stimulated expression of the MNC recruiting chemokine MCP-1. In biopsies, MNC infiltrates in decorin rich CP tissue showed a 300-fold upregulation of MCP-1 compared with decorin free peripheral blood, whereas no difference was found in basal MCP-1 expression in PBMC of patients versus donors. This effect was specific for MCP1-other inflammatory cytokines, such as interleukin 1beta and tumour necrosis factor alpha, were not affected. CONCLUSION: Decorin is a molecular marker of desmoplasia in CP, and excessive decorin may allow fibrotic masses to nourish and protract inflammation by deregulating the process of MNC accumulation and activation. These data provide a molecular basis for surgical resection of diseased tissue as a treatment option in CP.


Subject(s)
Pancreatitis, Chronic/metabolism , Proteoglycans/physiology , Adolescent , Adult , Aged , Blotting, Western , Cells, Cultured , Chemokine CCL2/blood , Decorin , Extracellular Matrix/metabolism , Extracellular Matrix Proteins , Female , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Pancreas/innervation , Proteoglycans/metabolism , Proteoglycans/pharmacology , RNA, Messenger/genetics , Recurrence , Reverse Transcriptase Polymerase Chain Reaction/methods , Up-Regulation
17.
Gut ; 55(4): 519-28, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16174661

ABSTRACT

BACKGROUND: Success of chemotherapy and alleviation of pain are frequently less than optimal in pancreatic cancer patients, leading to increasing interest in new pharmacological substances, such as vanilloids. Our study addressed the question of whether vanilloids influence pancreatic cancer cell growth, and if vanilloids could be used for pain treatment via the vanilloid 1 receptor (VR1) in pancreatic cancer patients. METHODS: In vitro, the effect of resiniferatoxin (vanilloid analogue) on apoptosis and cell growth in pancreatic cancer cells--either alone, combined with 5-fluorouracil (5-FU), or combined with gemcitabine--was determined by annexin V staining, FACS analysis, and MTT assay, respectively. VR1 expression was evaluated on RNA and protein level by quantitative polymerase chain reaction and immunohistochemistry in human pancreatic cancer and chronic pancreatitis. Patient characteristics--especially pain levels--were registered in a prospective database and correlated with VR1 expression. RESULTS: Resiniferatoxin induced apoptosis by targeting mitochondrial respiration and decreased cell growth in pancreatic cancer cells without showing synergistic effects with 5-FU or gemcitabine. Expression of VR1 was significantly upregulated in human pancreatic cancer and chronic pancreatitis. VR1 expression was related to the intensity of pain reported by cancer patients but not to the intensity of pain reported by patients with chronic pancreatitis. CONCLUSIONS: Resiniferatoxin induced apoptosis in pancreatic cancer cells indicates that vanilloids may be useful in the treatment of human pancreatic cancer. Furthermore, vanilloid might be a novel and effective treatment option for neurogenic pain in patients with pancreatic cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Diterpenes/therapeutic use , Pain/prevention & control , Pancreatic Neoplasms/drug therapy , Adult , Aged , Antimetabolites, Antineoplastic/therapeutic use , Apoptosis/drug effects , Cell Division/drug effects , Cell Line, Tumor , Chronic Disease , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Drug Synergism , Drug Therapy, Combination , Female , Fluorouracil/therapeutic use , Humans , Immunohistochemistry/methods , Male , Middle Aged , Mitochondria/drug effects , Oxidative Stress , Pancreas/chemistry , Pancreatic Neoplasms/complications , Pancreatitis/drug therapy , Prospective Studies , TRPV Cation Channels/analysis , Gemcitabine
18.
Cancer Biol Ther ; 4(7): 740-6, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15970685

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive malignancies, with an overall 5-year survival rate of less than 5%. Invasive tumor growth and early metastasis are two important reasons for this dismal prognosis. Osteopontin (OPN) is a secretory protein with a variety of functions, for example in cell adhesion and migration, inflammatory reaction and apoptosis. In this study the functional role of OPN in human pancreatic cancer and its potential use as a disease marker were analyzed. By real time quantitative PCR, there was a 2.2-fold and 1.6-fold increase of OPN mRNA in pancreatic cancers (n = 23) and chronic pancreatitis samples (n = 22), respectively, compared to normal pancreatic tissues (n = 20). Immunohistochemical analysis demonstrated OPN staining in 60% of the primary pancreatic tumors and in 72% of the lymph node and liver metastases. ELISA analysis of serum samples obtained from pancreatic cancer patients (n = 70), chronic pancreatitis patients (n = 12), and healthy donors (n = 20) showed a 1.6-fold increase in OPN serum levels in patients with tumors and a 1.9-fold increase in patients with chronic pancreatitis. Recombinant human OPN significantly increased the invasiveness of pancreatic cancer cells, without having any impact on cell proliferation. In addition, down regulation of OPN by specific siRNA molecules decreased pancreatic cancer cell invasion. In conclusion, OPN serum levels in pancreatic cancer and chronic pancreatitis patients are not significantly different, thereby restricting its role as a prognostic or follow-up marker. Our results do suggest, however, that blockade of OPN might be useful as a therapeutic approach to inhibit invasion and metastasis of pancreatic cancer cells.


Subject(s)
Pancreatic Neoplasms/pathology , Pancreatitis, Chronic/metabolism , Sialoglycoproteins/physiology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Biomarkers, Tumor/metabolism , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Case-Control Studies , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Lymphatic Metastasis/pathology , Neoplasm Invasiveness/pathology , Neoplasm Staging , Osteopontin , Pancreas/metabolism , Pancreatic Neoplasms/genetics , Pancreatitis, Chronic/genetics , Prognosis , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sialoglycoproteins/antagonists & inhibitors , Tumor Cells, Cultured
19.
J Gene Med ; 3(4): 326-37, 2001.
Article in English | MEDLINE | ID: mdl-11529662

ABSTRACT

BACKGROUND: The oncosuppressive properties of some autonomous parvoviruses such as H-1 virus, together with their low pathogenicity, make them attractive vectors for tumor-directed gene therapy. Indeed, it was recently shown that these viruses became endowed with an enhanced oncosuppressive activity after they had been engineered to deliver a recognized therapeutic transgene. This prompted us to use a parvoviral vector to analyse the antineoplastic capacity of MCP-3 (monocyte chemotactic protein-3), a CC chemokine which has a broad spectrum of target cells, and can thus be considered to be a promising candidate for cancer treatment. METHODS: We explored the use of a parvovirus H-1-based vector encoding human MCP-3 for its antitumor potential on human cervical carcinoma cells. HeLa cells were infected in vitro with the recombinant virus hH1/MCP-3 at a low multiplicity [1 replication unit (RU)/cell] and we investigated the effect of parvovirus-mediated MCP-3 transduction on tumor formation and growth upon implantation of HeLa cells in nude mice. RESULTS: Infection of HeLa cells with hH1/MCP-3 led to secretion of high levels of MCP-3 and to significant retardation of tumor growth in recipient mice, as compared with HeLa cells that were either buffer-treated or infected with a MCP-3-free vector. Tumors from hH1/MCP-3-infected HeLa cells were heavily infiltrated with activated macrophages and showed increased numbers of dendritic cells. In addition, activated natural killer (NK) cells were also recruited into MCP-3-transduced tumors. CONCLUSION: These observations indicate that parvovirus H-1-transduced MCP-3 is able to exert a significant antitumor activity which is mediated, at least in part, through macrophages and NK cells, under conditions in which activated T cells are lacking.


Subject(s)
Cytokines , Monocyte Chemoattractant Proteins/genetics , Parvovirus/genetics , Uterine Cervical Neoplasms/therapy , Animals , Chemokine CCL7 , Female , HeLa Cells , Humans , Mice , Mice, Nude , Monocyte Chemoattractant Proteins/pharmacokinetics , Monocyte Chemoattractant Proteins/therapeutic use , Plasmids , Recombinant Proteins/analysis , Transcription, Genetic , Transduction, Genetic , Transplantation, Heterologous
20.
J Pharmacol Exp Ther ; 298(3): 1172-8, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11504817

ABSTRACT

Exaggerated or inappropriate signaling by the platelet-derived growth factor receptor (PDGFR) tyrosine kinase has been implicated in a wide variety of diseases. Thus, a series of piperazinyl quinazoline compounds were identified as potent antagonists of the PDGFR by screening chemical libraries. An optimized analog, CT52923, was shown to be an ATP-competitive inhibitor that exhibited remarkable specificity when tested against other kinases, including all members of the closely related PDGFR family. The PDGFRs and stem cell factor receptor were inhibited with an IC(50) of 100 to 200 nM, while 45- to >200-fold higher concentrations of CT52923 were required to inhibit fms-like tyrosine kinase-3 and colony-stimulating factor-1 receptor, respectively. Other receptor tyrosine kinases, cytoplasmic tyrosine kinases, serine/threonine kinases, or members of the mitogen-activated protein kinase pathway were not significantly inhibited at 100- to 1000-fold higher concentrations. In addition, this compound also demonstrated specificity for inhibition of cellular responses. Platelet-derived growth factor-induced smooth muscle cell migration or fibroblast proliferation was found to be blocked by CT52923 with an IC(50) of 64 and 280 nM, respectively, whereas 50- to 100-fold higher concentrations were required to inhibit these responses when induced with fibroblast growth factor. To investigate the effect of CT52923 on PDGFR signaling, in vivo studies demonstrated that CT52923 could significantly inhibit neointima formation following carotid artery injury by oral administration in the rat. Therefore, PDGFR antagonism by CT52923 could be a viable strategy for the prevention of clinical restenosis or the treatment of other human diseases involving PDGFR signaling.


Subject(s)
Carotid Artery Injuries/pathology , Neovascularization, Pathologic/prevention & control , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Angioplasty, Balloon , Animals , CHO Cells , Cell Division/drug effects , Cell Movement/drug effects , Cricetinae , Cytoplasm/drug effects , Cytoplasm/enzymology , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Humans , Neovascularization, Pathologic/pathology , Phosphorylation , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Signal Transduction/drug effects , Thymidine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...