Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Am J Transplant ; 16(5): 1371-82, 2016 05.
Article in English | MEDLINE | ID: mdl-26602894

ABSTRACT

Donor alloantigen infusion induces T cell regulation and transplant tolerance in small animals. Here, we study donor splenocyte infusion in a large animal model of pulmonary transplantation. Major histocompatibility complex-mismatched single lung transplantation was performed in 28 minipigs followed by a 28-day course of methylprednisolone and tacrolimus. Some animals received a perioperative donor or third party splenocyte infusion, with or without low-dose irradiation (IRR) before surgery. Graft survival was significantly prolonged in animals receiving both donor splenocytes and IRR compared with controls with either donor splenocytes or IRR only. In animals with donor splenocytes and IRR, increased donor cell chimerism and CD4(+) CD25(high+) T cell frequencies were detected in peripheral blood associated with decreased interferon-γ production of leukocytes. Secondary third-party kidney transplants more than 2 years after pulmonary transplantation were acutely rejected despite maintained tolerance of the lung allografts. As a cellular control, additional animals received third-party splenocytes or donor splenocyte protein extracts. While animals treated with third-party splenocytes showed significant graft survival prolongation, the subcellular antigen infusion showed no such effect. In conclusion, minipigs conditioned with preoperative IRR and donor, or third-party, splenocyte infusions may develop long-term donor-specific pulmonary allograft survival in the presence of high levels of circulating regulatory T cells.


Subject(s)
Chimerism , Graft Survival/immunology , Isoantigens/immunology , Lung Transplantation , T-Lymphocytes, Regulatory/radiation effects , Animals , Female , Immunosuppression Therapy , Male , Models, Animal , Swine , Swine, Miniature , T-Lymphocytes, Regulatory/immunology , Tissue Donors , Transplantation Tolerance , Transplantation, Homologous , Whole-Body Irradiation
2.
Semin Cancer Biol ; 35 Suppl: S244-S275, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25865774

ABSTRACT

Cancer is a key health issue across the world, causing substantial patient morbidity and mortality. Patient prognosis is tightly linked with metastatic dissemination of the disease to distant sites, with metastatic diseases accounting for a vast percentage of cancer patient mortality. While advances in this area have been made, the process of cancer metastasis and the factors governing cancer spread and establishment at secondary locations is still poorly understood. The current article summarizes recent progress in this area of research, both in the understanding of the underlying biological processes and in the therapeutic strategies for the management of metastasis. This review lists the disruption of E-cadherin and tight junctions, key signaling pathways, including urokinase type plasminogen activator (uPA), phosphatidylinositol 3-kinase/v-akt murine thymoma viral oncogene (PI3K/AKT), focal adhesion kinase (FAK), ß-catenin/zinc finger E-box binding homeobox 1 (ZEB-1) and transforming growth factor beta (TGF-ß), together with inactivation of activator protein-1 (AP-1) and suppression of matrix metalloproteinase-9 (MMP-9) activity as key targets and the use of phytochemicals, or natural products, such as those from Agaricus blazei, Albatrellus confluens, Cordyceps militaris, Ganoderma lucidum, Poria cocos and Silybum marianum, together with diet derived fatty acids gamma linolenic acid (GLA) and eicosapentanoic acid (EPA) and inhibitory compounds as useful approaches to target tissue invasion and metastasis as well as other hallmark areas of cancer. Together, these strategies could represent new, inexpensive, low toxicity strategies to aid in the management of cancer metastasis as well as having holistic effects against other cancer hallmarks.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/genetics , Cadherins/genetics , Humans , Neoplasm Invasiveness/genetics , Neoplasm Metastasis , Neoplasms/pathology , Signal Transduction/drug effects , Tight Junctions/drug effects , Tight Junctions/genetics
3.
Internist (Berl) ; 56(2): 127-8, 130-6, 2015 Feb.
Article in German | MEDLINE | ID: mdl-25636955

ABSTRACT

The incidence of obesity in the western world has increased dramatically during recent decades. Epidemiological data suggest that obesity is associated with an increased risk of several but not all types of cancers, with clear sex-specific differences. The underlying mechanisms are still a matter of debate. This review focuses on the potential factors linking obesity to cancer. Current experimental evidence suggests that insulin resistance and a chronic, subclinical inflammation in the visceral fat are the major metabolic events causing alterations in the levels of insulin, glucose, free fatty acids, insulin-like growth factor 1 (IGF-1) and 2, adipose tissue-derived proinflammatory cytokines and other bioactive molecules, such as adipokines (e.g. leptin and adiponectin), vascular endothelial growth factor (VEGF), sex hormones, gut microbiota and secondary bile acids. All these factors may act directly or indirectly on the tumor microenvironment to drive tumor progression via stimulation of cell survival/antiapoptosis, cell proliferation, angiogenesis and invasion/metastasis of the cancer cells. Therapeutic strategies that target dysfunctional or inflamed fat and have been shown to benefit patients include bariatric surgery, while other cell or hormone-directed interventions, such as conversion of visceral fat macrophages to an anti-inflammatory M2 phenotype or the pharmacological modulation of serum adipokine levels are still theoretical and need to be clinically evaluated for their ability to successfully treat or prevent obesity-related cancers.


Subject(s)
Cytokines/immunology , Neoplasms/epidemiology , Neoplasms/immunology , Obesity/epidemiology , Obesity/immunology , Tumor Microenvironment/immunology , Causality , Comorbidity , Humans , Models, Immunological , Risk Factors
5.
Adv Biochem Eng Biotechnol ; 130: 89-103, 2013.
Article in English | MEDLINE | ID: mdl-22936399

ABSTRACT

During the last decade it was realized that stem cell-based therapies hold an enormous therapeutic potential, improving the life of patients with conditions ranging from neurodegenerative and traumatic diseases to regenerative medicine requiring replacement of complex structures such as bones and teeth. Based on their ability to regenerate and/or repair damaged tissue and eventually restore organ function, multiple types of stem/progenitor cells have been discovered. In the field of periodontal regeneration and tooth engineering, several types of adult multipotent mesenchymal stem cells from various sources are currently being investigated. These include the bone marrow stromal stem cells (BMSSCs), adipose-derived stromal cells (ADSCs), dental pulp stem cells (DPSCs), dental follicle stem cells (DFSCs), stem cells from human exfoliated deciduous teeth (SHEDs), stem cells from the apical papilla (SCAP), periodontal ligament stem cells (PDLSCs), alveolar bone proper-derived stem cells, and gingival stem cells. The potential of these different MSCs as precursors for regenerative purposes in the dental field is discussed in this chapter.

7.
Oncogene ; 26(33): 4850-62, 2007 Jul 19.
Article in English | MEDLINE | ID: mdl-17297450

ABSTRACT

In the present study, we have analysed the effects of transforming growth factor-beta (TGF-beta) signaling on the growth behavior of pancreatic carcinoma cells in vitro and on their tumorigenicity in vivo. Ectopic expression of dominant-negative mutants of the TGF-beta type II receptor or type I receptor/activin receptor-like kinase 5 (ALK5) in TGF-beta-sensitive pancreatic ductal adenocarcinoma PANC-1 cells prevented the TGF-beta-induced activation of transfected Smad-responsive reporter genes and growth arrest. The growth-inhibitory effect was mimicked by stable expression of kinase-active ALK5 (ALK5-T204D), and was dependent on ALK5's ability to activate Smad signaling, as a ALK5-derived mutant with an intact kinase domain but deficient in its ability to activate Smads (RImL45) failed to suppress proliferation in the absence of added TGF-beta. Moreover, this mutant often displayed opposite effects to those of ALK5-TD and blocked various ligand-induced responses in vitro, indicating that it acts in a dominant-negative fashion to inhibit endogenous wild-type receptors. ALK5-TD-, but not RImL45-TD-transduced cells underwent epithelial-to-mesenchymal transition, exhibited a higher ratio of thrombospondin-1 to vascular endothelial growth factor-A expression and upregulated various metastasis-associated genes. Upon orthotopic transplantation of PANC-1 clones into immunodeficient mice, ALK5-TD, but not RImL45-TD, greatly reduced tumor size and induced the formation of liver metastases in otherwise non-metastatic PANC-1 cells. These results suggest a causal, dominant role for the endogenous Smad2/3 signaling pathway in the tumor suppressor and prometastatic activities of TGF-beta in pancreatic tumor cells.


Subject(s)
Activin Receptors, Type I/physiology , Adenocarcinoma/pathology , Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/pathology , Receptors, Transforming Growth Factor beta/physiology , Activin Receptors, Type I/genetics , Activin Receptors, Type I/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression/drug effects , Humans , Immunoblotting , Mice , Mice, SCID , Mutation , Neoplasm Metastasis , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Serine-Threonine Kinases , Rats , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Smad Proteins/metabolism , Transfection , Transforming Growth Factor beta/pharmacology , Tumor Burden
8.
Am J Transplant ; 6(5 Pt 1): 947-58, 2006 May.
Article in English | MEDLINE | ID: mdl-16611330

ABSTRACT

Recently, we generated cells with multipotent properties from blood monocytes that in vitro differentiate into various somatic cell types. This experimental study investigated whether these programmable cells of monocytic origin (PCMO) succeed to restore left ventricular function after myocardial infarction (MI). PCMO were generated from monocytes by exposition to RPMI medium containing M-CSF and IL-3 for 6 days. MI was induced in female Lewis rats ligating the left coronary artery. PCMO of male Lewis donors were injected either intramyocardially (i.my.) or intravenously (i.v.) 24 h or 6 days post-infarction. Hemodynamic assessment after 60 days demonstrated significant improvement of left ventricular function following i.my. transplantation of PCMO as well as early (24 h post-infarction) i.v. application while nonmodulated monocytes failed to restore heart function. The Y-chromosome-specific SRY gene of male donor PCMO was detected exclusively in infarcted hearts of animals, which demonstrated improved cardiac function. Subdivision of infarcted hearts by microdissection localized the SRY gene-containing department to the left ventricle adjacent to the infarcted area whereas the right ventricle remained negative. Successful generation of PCMO in access numbers allows their autologous use as a new additive treatment for early restoration of cardiac function after MI.


Subject(s)
Heart Function Tests , Monocytes/transplantation , Myocardial Infarction/therapy , Stem Cell Transplantation , Ventricular Function, Left , Animals , Capillaries/pathology , Coronary Circulation , Disease Models, Animal , Echocardiography , Female , Hemodynamics , Male , Polymerase Chain Reaction , Rats , Rats, Inbred Lew , Vascular Endothelial Growth Factor A/genetics , Y Chromosome
9.
Acta Gastroenterol Belg ; 68(4): 461-5, 2005.
Article in English | MEDLINE | ID: mdl-16433004

ABSTRACT

There is currently great excitement and expectation concerning the differential potential of adult stem cells or adult cells with capacity of differentiation. As the body of work concerning transdifferentiation of somatic stem cells and bone marrow derived stem cells grows, the number of critics increases steadily questioning the reliability of reported findings. So scientists are now challenged more and more to prove that resulting differentiated somatic cells originated from somatic adult stem cell through a transdifferentiation process. Phenomenons such as fusion of cells have to be ruled out and the origin of the differentiated cell has to be determined by specific techniques i.e. in situ hybridisation. Cellular mimicry through uptake of specific factors out of the medium is questioned to be the reason for cells staining positive for Insulin. Some multipotent adult stem cells can cross lineage boundaries and differentiate into somatic cells of other lineages after being relocated. Bone marrow cells have been described to have the greatest plasticity among adult stem cells regenerating damaged liver or myocardium. It has been proposed that the differentiation of bone marrow derived adult stem cells occurs naturally even in healthy organs as a physiologic process of tissue-regeneration. Others believe that organ damage is essential to induce transdifferentiation by release of organ specific microenvironmental factors. We here try to constitute necessary data which should be demonstrated to give substantial evidence for transdifferentiation of newly characterized cells including exclusion of fusion, phagocytosis or DNA uptake, description of the outset cell, differentiation into all three germ layers and functional parameters.


Subject(s)
Metabolism, Inborn Errors/therapy , Multipotent Stem Cells/transplantation , Stem Cell Transplantation/methods , Transplantation Immunology/physiology , Adult , Animals , Female , Germany , Graft Rejection , Graft Survival , Humans , Male , Metabolism, Inborn Errors/diagnosis , Mice , Molecular Mimicry/physiology , Prognosis , Risk Factors , Transplantation, Autologous
10.
Br J Cancer ; 89(9): 1714-21, 2003 Nov 03.
Article in English | MEDLINE | ID: mdl-14583775

ABSTRACT

Inability to die by apoptosis is one of the reasons for the deregulated growth of tumour cells and the frequently observed failure of chemotherapy. In this study we thought to identify the common and functionally important characteristics responsible for the apoptosis resistance of pancreatic tumour cells. We analysed cell surface expression level of death receptors CD95 and TRAIL-R1-4 as well as the expression profile of sixteen apoptosis-relevant proteins in five pancreatic carcinoma cell lines Capan1, Colo357, PancTuI, Panc89 and Panc1. These data were evaluated in the context of sensitivity towards anti-CD95 and TRAIL-mediated apoptosis. Here we report that except for resistant Panc1 cells, which only marginally expressed CD95, all other cell lines showed comparable levels of CD95 and TRAIL receptors irrespectively of their apoptotic phenotype. Interestingly, we found that the elevated expression of FLIP, Bcl-x(L) and IAP in parallel with a downregulation of FADD and Bid was common for the resistant cell lines. Consequently, stable overexpression of XIAP, Bcl-x(L) or dominant negative FADD in sensitive cells significantly reduced the death receptor mediated apoptosis while the overexpression of Bid rendered the resistant cells sensitive.


Subject(s)
Adenocarcinoma/genetics , Apoptosis/genetics , Biomarkers, Tumor/analysis , Cell Transformation, Neoplastic/genetics , Pancreatic Neoplasms/genetics , Adenocarcinoma/metabolism , Apoptosis Regulatory Proteins , Blotting, Western , Caspases/biosynthesis , Cell Line, Tumor/physiology , Enzyme Activation , Flow Cytometry , Humans , Membrane Glycoproteins/biosynthesis , Pancreatic Neoplasms/metabolism , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/biosynthesis , fas Receptor/biosynthesis
11.
Stem Cells ; 21(4): 428-36, 2003.
Article in English | MEDLINE | ID: mdl-12832696

ABSTRACT

The in vitro differentiation of mouse embryonic stem cells into different somatic cell types such as neurons, endothelial cells, or myocytes is a well-established procedure. Long-term culture of rat embryonic stem cells is known to be hazardous, and attempts to differentiate these cells in vitro so far have been unsuccessful. We herein describe stable long-term culture of an alkaline phosphatase-positive rat embryonic stem cell-like cell line (RESC) and its differentiation into neuronal, endothelial, and hepatic lineages. RESCs were characterized by typical growth in single cells as well as in embryoid bodies when cultured in the presence of leukemia inhibitory factor. RESC expressed stage-specific-embryonic antigen-1 and the major histocompatibility complex class I molecule. For neuronal differentiation, cells were incubated with medium containing 10(-6) M retinoic acid for 14 days. For endothelial differentiation, RESCs were grown on Matrigel for 14 days, and for induction of hepatocyte-specific antigen expression, RESCs were grown in medium supplemented with fibroblast growth factor-4. Differentiated cells exhibited typical morphological changes and expressed neuronal (nestin, mitogen-activated protein-2, synaptophysin), glial (S100, glial fibrillary acid protein), endothelial (panendothelial antibody, CD31) and hepatocyte-specific (alpha-fetoprotein [alphaFP], albumin, alpha-1-antitrypsin, CK18) antigens. In addition, expression of hepatocyte-specific genes (alphaFP, transthyretin, carbamoyl-phosphate synthetase, and coagulation factor-2) was detected by reverse transcription polymerase chain reaction. We were able to culture RESCs under stable, long-term conditions and to initiate programmed differentiation of RESCs to endothelial, neuronal, glial, and hepatic lineages in the rat species.


Subject(s)
Endothelium, Vascular/cytology , Neuroglia/cytology , Neurons/cytology , Stem Cells/cytology , Animals , Cell Differentiation , Cell Division , Cell Line , Cell Lineage , Collagen/pharmacology , Drug Combinations , Embryo, Mammalian/cytology , Endothelial Cells/cytology , Epithelial Cells/metabolism , Fibroblast Growth Factor 4 , Fibroblast Growth Factors/metabolism , Genes, MHC Class I , Hepatocytes/metabolism , Immunohistochemistry , Laminin/pharmacology , Liver/metabolism , Phenotype , Proteoglycans/pharmacology , Proto-Oncogene Proteins/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tretinoin/pharmacology
12.
J Cell Sci ; 114(Pt 15): 2735-46, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11683408

ABSTRACT

In this study we investigated the functional role of FAP-1 as a potential inhibitor of CD95 (Fas, APO-1)-mediated apoptosis in pancreatic cancer cells. Stable transfection of the CD95-sensitive, FAP-1-negative cell line Capan-1 with an FAP-1 cDNA resulted in a strongly decreased sensitivity to CD95-induced apoptosis, as measured by DNA fragmentation and caspase-3 activity. Inhibition of cellular protein tyrosine phosphatases with orthovanadate dose-dependently increased CD95-induced apoptosis in CD95-resistant FAP-1-positive Panc89 and Capan-1-FAP-1 cells almost to the level seen in wild-type Capan-1 cells. Blocking the CD95/FAP-1 interaction in Panc89 cells by cytoplasmic microinjection of a synthetic tripeptide mimicking the C terminus of CD95 resulted in a mean 5.5-fold increase in apoptosis compared to cells that received a control peptide. Using confocal laser scanning microscopy we show that in Panc89 cells FAP-1 is mainly associated with the Golgi complex and with peripheral vesicles. FAP-1 displayed enhanced colocalization with CD95 upon CD95 stimulation in the Golgi complex but not in surface-associated vesicles. This correlated with a decrease in plasma membrane staining for CD95 as determined by FACS analysis. Inhibition of Golgi anterograde transport by brefeldin A abolished the anti-CD95-induced colocalization of FAP-1 and CD95 as well as the decrease in cell-surface-associated CD95. Finally, we demonstrate by immunohistochemistry that FAP-1 is strongly expressed in tumor cells from pancreatic carcinoma tissues. Taken together, these results show that FAP-1 can protect pancreatic carcinoma cells from CD95-mediated apoptosis, probably by preventing anti-CD95-induced translocation of CD95 from intracellular stores to the cell surface.


Subject(s)
Adenocarcinoma , Apoptosis/immunology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Pancreatic Neoplasms , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , fas Receptor/metabolism , Brefeldin A/pharmacology , Carrier Proteins/analysis , Cytoplasmic Vesicles/chemistry , Cytoplasmic Vesicles/metabolism , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Golgi Apparatus/chemistry , Golgi Apparatus/metabolism , Humans , Jurkat Cells , Peptide Fragments/pharmacology , Protein Synthesis Inhibitors/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 13 , Protein Tyrosine Phosphatases/analysis , Protein Tyrosine Phosphatases/antagonists & inhibitors , fas Receptor/analysis
13.
Oncogene ; 20(31): 4258-69, 2001 Jul 12.
Article in English | MEDLINE | ID: mdl-11464292

ABSTRACT

The molecular alterations in tumour cells leading to resistance towards apoptosis induced by CD95 and TRAIL-receptors are not fully understood. We report here that the stimulation of the CD95- and TRAIL-resistant human pancreatic adenocarcinoma cell line PancTuI with an agonistic anti-CD95 antibody or TRAIL resulted in activation of protein kinase C and NF-kappaB. Inhibition of protein kinase C by Gö6983 sensitized these cells to apoptotic challenges and strongly diminished activation of NF-kappaB by anti-CD95 and TRAIL. Similarly, inhibition of NF-kappaB by MG132 or by transient transfection with a dominant negative mutant of IkappaBalpha restored the responsiveness of PancTuI cells to both death ligands. In the CD95 and TRAIL-sensitive cell line Colo357 the induction of protein kinase C and NF-kappaB following activation of CD95 and TRAIL-R was very moderate compared with PancTuI cells. However, pre-incubation of these cells with PMA strongly reduced their apoptotic response to anti-CD95 and TRAIL. Taken together, we show that activation of protein kinase C operates directly in a death receptor-dependent manner in PancTuI cells and protect pancreatic tumour cells from anti-CD95 and TRAIL-mediated apoptosis by preventing the loss DeltaPsim and Cytochrome c release as well as by induction of NF-kappaB.


Subject(s)
Adenocarcinoma/pathology , Apoptosis/physiology , Membrane Glycoproteins/physiology , NF-kappa B/metabolism , Pancreatic Neoplasms/pathology , Protein Kinase C/metabolism , Tumor Necrosis Factor-alpha/physiology , fas Receptor/physiology , Adenocarcinoma/enzymology , Adenocarcinoma/metabolism , Apoptosis Regulatory Proteins , Enzyme Activation , Humans , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/metabolism , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured
14.
FEBS Lett ; 494(3): 196-200, 2001 Apr 13.
Article in English | MEDLINE | ID: mdl-11311240

ABSTRACT

The early response gene IEX-1 modulates apoptosis and cell growth in a poorly defined fashion. Here, we describe the effect of hammerhead ribozymes specifically disrupting IEX-1 expression in 293 cells. Compared to vector control, 293 cells exhibit a reduced growth rate and a slowed cell cycle progression, when stably transfected with a concatemeric ribozyme construct. In addition, these 293 cells were much less sensitive to apoptosis induced by an activating Fas/CD95 antibody or by the anti-cancer drugs etoposide and doxorubicin. By modulating the cell cycle, IEX-1 might be part of a growth signal if favourable growth conditions prevail, whereas under unfavourable conditions, i.e. death receptor activation, IEX-1 facilitates apoptosis.


Subject(s)
Immediate-Early Proteins/metabolism , Membrane Glycoproteins/metabolism , Neoplasm Proteins , RNA, Catalytic/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Base Sequence , Cell Division/drug effects , Cell Line , Doxorubicin/pharmacology , Etoposide/pharmacology , Humans , Immediate-Early Proteins/biosynthesis , Immediate-Early Proteins/genetics , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Membrane Proteins , RNA, Catalytic/chemistry , RNA, Catalytic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Templates, Genetic , Transfection , fas Receptor/metabolism
15.
Virchows Arch ; 439(6): 798-802, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11787853

ABSTRACT

The K-ras, p53, p16 and DPC4 genes are among those most frequently altered in pancreatic ductal carcinoma. We analyzed 22 cell lines for alterations in these genes by direct sequence analysis and methylation-specific polymerase chain reaction. These cell lines showed mutations in K-ras and p53 at frequencies of 91% and 95%, respectively. Alterations in p16INK4a were found in all cases and included nine homozygous deletions, seven mutations and promoter methylation in six cases. Eight cell lines (36%) had an alteration of DPC4, including one mutation and seven homozygous deletions. The most typical mutational profile involved K-ras, p53, and p16INK4a, concurrently aberrated in 20 cases (91%). Eight cell lines had alterations in all four genes. Inactivation of DPC4 was always accompanied by alteration of all of the other three genes. This comprehensive data regarding the cumulative genetic alterations in pancreatic carcinoma cell lines will be of great value for studies involving drug sensitivity or resistance that may be associated with inactivation of a particular gene or molecular pathway.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , DNA-Binding Proteins/genetics , Genes, p16 , Genes, p53 , Genes, ras , Pancreatic Neoplasms/genetics , Trans-Activators/genetics , DNA Mutational Analysis , DNA, Neoplasm/analysis , Humans , Mutation , Polymerase Chain Reaction , Smad4 Protein , Tumor Cells, Cultured
16.
Oncogene ; 19(48): 5477-86, 2000 Nov 16.
Article in English | MEDLINE | ID: mdl-11114725

ABSTRACT

In this study we sought to clarify the role of the proapoptotic potential of mitochondria in the death pathway emanating from the TRAIL (APO-2L) and CD95 receptors in pancreatic carcinoma cells. We focused on the role of the Bcl-2 family member Bcl-XL, using three pancreatic carcinoma cell lines as a model system, two of which have high (Panc-1, PancTuI) and one has low (Colo357) Bcl-XL expression. In these cell lines, the expression of Bcl-XL correlated with sensitivity to apoptosis induced by TRAIL or anti-CD95. Flow cytometric analysis revealed cell surface expression of TRAIL-R1 and TRAIL-R2 on PancTuI and Colo357, and TRAIL-R2 on Panc-1 cells. In Colo357 cells retrovirally transduced with Bcl-XL, caspase-8 activation in response to treatment with TRAIL or anti-CD95 antibody was not different from parental cells and EGFP-transfected controls, however, apoptosis was completely suppressed as measured by the mitochondrial transmembrane potential deltapsim, caspase-3 activity (PARP cleavage) and DNA-fragmentation. Inhibition of Bcl-XL function by overexpression of Bax or administration of antisense oligonucleotides against Bcl-XL mRNA resulted in sensitization of Panc-1 cells to TRAIL and PancTuI cells to anti-CD95 antibody-induced cell death. The results show that Bcl-XL can protect pancreatic cancer cells from CD95- and TRAIL-mediated apoptosis. Thus, in these epithelial tumour cells the mitochondrially mediated 'type II' pathway of apoptosis induction is not only operative regarding the CD95 system but also regarding the TRAIL system.


Subject(s)
Adenocarcinoma/pathology , Apoptosis/physiology , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/physiology , Receptors, Tumor Necrosis Factor/physiology , fas Receptor/physiology , Adenocarcinoma/metabolism , Humans , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/biosynthesis , Signal Transduction/physiology , Transfection , Tumor Cells, Cultured , bcl-2-Associated X Protein , bcl-X Protein
17.
Ann N Y Acad Sci ; 880: 243-51, 1999 Jun 30.
Article in English | MEDLINE | ID: mdl-10415870

ABSTRACT

Malignancies have developed several strategies to evade immune surveillance. We have investigated pancreatic cancer cell lines and pancreatic cancer surgical specimens to evaluate possibilities of tumor escape in the Fas system, and local immune suppression. Despite Fas expression the majority of cell lines was resistant to Fas-mediated apoptosis. The Fas-associated phosphatase-1 is a strong candidate to confer Fas resistance in pancreatic cancer cells. In addition, all investigated pancreatic cancer cell lines and cancer specimens expressed Fas ligand. Fas ligand was functional in cancer cell lines as shown by coculture assays of pancreatic cancer cell lines with Jurkat cells as targets. Additional local immune suppression was demonstrated by loss of T-cell receptor/CD3-zeta chain of pancreatic cancer infiltrating T-lymphocytes. We conclude that these tumor escape mechanisms may contribute to the poor prognosis of pancreatic cancer but also represent targets for new treatment modalities.


Subject(s)
Adenocarcinoma/immunology , Pancreatic Neoplasms/immunology , Adenocarcinoma/pathology , Apoptosis , Carrier Proteins , Fas Ligand Protein , Humans , Jurkat Cells , Lymphocyte Activation , Membrane Glycoproteins , Pancreatic Neoplasms/pathology , Protein Phosphatase 1 , Protein Tyrosine Phosphatase, Non-Receptor Type 13 , Protein Tyrosine Phosphatases , Receptor-CD3 Complex, Antigen, T-Cell , T-Lymphocytes, Cytotoxic , Tumor Cells, Cultured , fas Receptor
18.
Int J Oncol ; 14(1): 93-101, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9863014

ABSTRACT

It is generally assumed that TGFbeta induces cell cycle arrest through the cooperative action of cell cycle inhibitors p15, p27 and p21. Here, we found that several pancreatic carcinoma cell lines exert TGFbeta-induced negative growth control in spite of the loss of p15 and p16 expression. In these cell lines, TGFbeta-induced growth control correlates with the upregulation of the p21 protein and active pRb expression. Conversely, cells without p21 and/or pRb expression are resistant to TGFbeta -induced growth inhibition. Moreover, overexpression of p21 in the p21-deficient cell line Panc Tu1 leads to growth arrest. Thus, TGFbeta-induced growth control correlates with p21 expression and pRb status independent of p15 and/or p16 expression.


Subject(s)
Cell Cycle Proteins , Cyclins/analysis , Growth Inhibitors/pharmacology , Retinoblastoma Protein/physiology , Transcription Factors/analysis , Transforming Growth Factor beta/pharmacology , Tumor Suppressor Proteins , Cyclin-Dependent Kinase Inhibitor p15 , Cyclin-Dependent Kinase Inhibitor p16/analysis , Cyclin-Dependent Kinase Inhibitor p21 , G1 Phase , Humans , Pancreatic Neoplasms/pathology , Tumor Cells, Cultured
19.
J Biol Chem ; 273(44): 29230-40, 1998 Oct 30.
Article in English | MEDLINE | ID: mdl-9786935

ABSTRACT

In this study we demonstrate that the gene encoding the small leucine-rich proteoglycan biglycan is expressed in human myometrial tissue and in the human leiomyosarcoma cell line SK-UT-1. Treatment of SK-UT-1 cells with forskolin or 8-bromo-cAMP strongly increased biglycan mRNA and this effect was transcriptional as shown by transient transfection experiments with biglycan promoter-luciferase reporter fusion genes. The cAMP-mediated induction of the transfected biglycan promoter in SK-UT-1 cells was abolished by coexpression of a specific protein kinase A inhibitor, and was mimicked by overexpression of the catalytic subunit (Cbeta) of protein kinase A. By 5' deletion analysis, part of the cAMP response was localized to the segment from residues -78 to -46 of the biglycan promoter. This region conferred strong cAMP responsiveness to a heterologous promoter. Electrophoretic mobility shift and antibody supershift assays identified two specific complexes that contained nuclear proteins antigenically related to the ubiquitous transcription factors Sp1 and Sp3, respectively. The binding site of these proteins was mapped to a CT-rich sequence extending from -59 to -49 in the biglycan promoter. Mutating this sequence eliminated complex formation and markedly reduced basal and cAMP-dependent promoter activity of transfected reporter genes. In vitro binding studies using recombinant Sp1 revealed that the nuclear factor binding to the CT element was not Sp1 but a Sp1-like protein(s). Western blot analysis of SK-UT-1 nuclear proteins confirmed expression of Sp3, Sp1 and nuclear proteins that crossreacted with Sp1 antibody but according to their molecular weight were not Sp1. These results indicate that all cAMP-dependent as well as some basal biglycan transcription in SK-UT-1 cells is mediated through activated protein kinase A and that both functions are conferred at the promoter level through the interaction of Sp1-like/Sp3 factors with the CT element at -59 in the biglycan promoter.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , DNA-Binding Proteins/metabolism , Leiomyosarcoma/genetics , Proteoglycans/genetics , Sp1 Transcription Factor/metabolism , Transcription Factors/metabolism , Transcription, Genetic , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Base Sequence , Biglycan , Colforsin/pharmacology , Cyclic AMP/pharmacology , DNA Primers , DNA, Complementary , Extracellular Matrix Proteins , Female , Gene Expression Regulation/drug effects , Humans , Leiomyosarcoma/pathology , Myometrium/drug effects , Myometrium/metabolism , Promoter Regions, Genetic , Protein Binding , Sp3 Transcription Factor , Transfection , Tumor Cells, Cultured
20.
Cancer Res ; 58(8): 1741-9, 1998 Apr 15.
Article in English | MEDLINE | ID: mdl-9563493

ABSTRACT

The Fas system, comprising the Fas receptor (Fas, CD95, APO-1) and its ligand, Fas ligand (FasL), is a central mediator of programmed cell death in various physiological and pathological processes. Recent evidence indicated that tumor cells can exploit this system to their benefit in the dialogue with the host immune system. We have shown that all human pancreatic adenocarcinoma cell lines tested by fluorescence-activated cell sorting analysis (6 of 6) and immunocytochemistry (12 of 12) were positive for Fas expression, as were normal and malignant duct cells in pancreatic tissue sections. However, despite Fas expression, pancreatic tumor cells have become largely resistant toward recombinant FasL- or anti-APO-1 agonistic antibody-induced apoptosis. This resistance correlated with high levels in pancreatic tumor cells of mRNA for FAP-1, a Fas-associated phosphatase that can block the apoptotic function of Fas. Using a variety of methodological approaches, we also present evidence for the production of FasL by pancreatic tumor cells because 6 of 6 pancreatic tumor cell lines were found to contain FasL mRNA as well as the Mr 40,000 and Mr 26,000 forms of the FasL protein. Likewise, pancreatic tissue revealed FasL-specific immunostaining in pancreatic tumor cells but not in the surrounding stroma. In coculture experiments, pancreatic tumor cells displayed a cytotoxic effect toward the Fas-sensitive Jurkat T-cell line, which could be inhibited by a FasL-specific neutralizing antibody. Together, these results support the recently proposed "counterattack model" for local deletion of tumor-reactive T-cells by tumor cell-derived FasL.


Subject(s)
Adenocarcinoma/metabolism , Apoptosis , Membrane Glycoproteins/metabolism , Pancreatic Neoplasms/metabolism , fas Receptor/metabolism , Antibodies/pharmacology , Carrier Proteins/metabolism , Cell Membrane/metabolism , Coculture Techniques , Culture Media, Conditioned/chemistry , Cytoplasm/metabolism , Fas Ligand Protein , Flow Cytometry , Humans , Immunoblotting , Immunohistochemistry , Jurkat Cells , Membrane Glycoproteins/pharmacology , Polymerase Chain Reaction , Protein Tyrosine Phosphatase, Non-Receptor Type 13 , Protein Tyrosine Phosphatases/metabolism , RNA, Messenger/analysis , Recombinant Proteins/pharmacology , Tumor Cells, Cultured , fas Receptor/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...