Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
1.
Gene Ther ; 20(9): 939-48, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23552473

ABSTRACT

DNA vaccines are attractive candidates for tumor immunotherapy. However, the potential of DNA vaccines in treating established malignant lesions has yet to be demonstrated. Here we demonstrate that transient alteration of either intratumoral or intradermal (ID) chemotactic gradients provide a favorable milieu for DNA vaccine-mediated activation of tumor-specific immune response in both prophylactic and therapeutic settings. Specifically, we show that priming of established B16 ID melanoma lesions via forced intratumoral expression of CCL21 boosted DNA vaccination-dependent systemic cytotoxic immune response leading to the regression of tumor nodules. In this setting, application of CCL20 was not effective likely due to the engagement of the regulatory T cells. However, priming of the skin at DNA vaccine administration sites outside the tumor bed with both CCL20 and CCL21 chemokines along with structural modifications of the DNA vaccine significantly improved vaccine efficacy. This optimized ID vaccination regimen led to the inhibition of distant established melanomas and prolonged tumor-free survival of mice observed in 60% of vaccinated animals with complete tumor remission in 30%. These effects were mediated by extranodal priming and activation of T cells at vaccine administration sites and progressive accumulation of systemic antigen-specific cytotoxic T cells (CTLs) on successive vaccinations. These results underscore the potential of chemokine-enhanced DNA vaccination to mount therapeutic immune response against established tumors.


Subject(s)
Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Chemokine CCL20/immunology , Chemokine CCL21/immunology , Melanoma, Experimental/therapy , Vaccines, DNA/immunology , Vaccines, DNA/therapeutic use , Animals , Cancer Vaccines/administration & dosage , Cell Line, Tumor , Chemokine CCL20/genetics , Chemokine CCL20/metabolism , Chemokine CCL21/genetics , Chemokine CCL21/metabolism , Genetic Therapy , Immunotherapy , Lymphocyte Activation , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/prevention & control , Mice , Mice, Inbred C57BL , Skin/immunology , Skin/metabolism , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , Skin Neoplasms/therapy , T-Lymphocytes/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccines, DNA/metabolism
2.
IET Syst Biol ; 2(1): 1-4, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18248079

ABSTRACT

Receptor tyrosine kinases (RTKs) are transmembrane allosteric enzymes: binding of ligand growth factors to their ectodomains stimulates a cytoplasm-facing tyrosine kinase activity, which initiates a plethora of cellular processes. The enormous complexity of RTK signalling, along with rich involvement in pathologies (e.g. cancer and diabetes), motivated the establishment of the international, multi-disciplinary RTK consortium (http://www.rtkconsort.org/) in 2005. In collaboration with the British Society for Proteome Research and the European Bioinformatics Institute, the Consortium held on July 23rd and 24th a Workshop on Proteomics and Phosphoproteomics of RTK Signalling Networks (Hinxton Hall Conference Centre, Cambridge, UK). As highlighted below, systems control (a layered web of regulatory loops summarised in Fig.1) emerged throughout the workshop as a common theme of many presentations.


Subject(s)
Feedback/physiology , Gene Expression Regulation/physiology , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Protein Processing, Post-Translational/physiology , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/physiology , Animals , Humans
3.
Clin Pharmacol Ther ; 82(1): 70-80, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17495877

ABSTRACT

In vivo studies represent an essential step in drug development and currently rely largely on mice, yet limitations of mammalian models motivated the search for complementary vertebrate model systems. This review focuses on zebrafish, Danio rerio, as a facile model system to study human disease and drug responses. Zebrafish are particularly suited for this purpose because they represent a vertebrate species, their genome is sequenced, and a large number of synchronously developing, transparent embryos can be produced. Zebrafish embryos are permeable to drugs and can easily be manipulated using well-established genetic and molecular approaches. Here, we summarize recent work on drug discovery and toxicity in zebrafish embryos. In addition, we provide a synopsis of current efforts to establish disease models in zebrafish focusing on neoplasia. The results of these studies highlight the potential of zebrafish as a viable addition to established animal models by offering medium and, potentially, high throughput capabilities.


Subject(s)
Drug Design , Drug Evaluation, Preclinical/methods , Models, Animal , Toxicity Tests/methods , Zebrafish/embryology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinogenicity Tests/methods , Carcinogens/toxicity , Cell Transformation, Neoplastic/drug effects , DNA Damage , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/pathology , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mutagenicity Tests/methods , Mutagens/toxicity , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/embryology , Reproducibility of Results , Zebrafish/genetics
4.
Exp Dermatol ; 12(2): 191-7, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12702148

ABSTRACT

Recently, we identified several missense mutations of the connexin gene GJB3 encoding connexin 31 (Cx31) in erythrokeratodermia variabilis (EKV), an autosomal dominant skin disorder. These mutations include G12D, which replaces a conserved glycine residue in the amino-terminus of Cx31 and is associated with a severe EKV phenotype. In contrast, the biologic relevance of the GJB3 sequence variant R32W located in the first transmembrane domain of Cx31 is disputed. To examine the effects of these sequence variants on Cx31 biogenesis and gap junction activity we expressed wild type and mutant Cx31-Flag constructs in HeLa cells. Using immunostaining, all expression variants were detected in the cytoplasm and in a punctate pattern at the cell surface, indicating that G12D and R32W did not interfere with either protein synthesis or transport to the cell membrane. Similarly, oligomerization into hemichannels appeared not impaired when expressing either Cx31 mutant as assessed by size exclusion chromatography, immunoblotting and immunostaining. However, dye transfer experiments and monitoring of intracellular calcium levels in response to serum stimulation revealed that G12D-Cx31 did not form functional gap junction channels, probably due to incorrect assembly or altered properties of Cx31 channels. In contrast, intercellular coupling between cells expressing R32W-Cx31 was comparable to that of wtCx31, suggesting that R32W is a functionally inconsequential polymorphism of Cx31.


Subject(s)
Connexins/genetics , Connexins/physiology , Base Sequence , Calcium Signaling , Connexins/chemistry , DNA, Complementary/genetics , Gap Junctions/physiology , Genetic Variation , HeLa Cells , Humans , In Vitro Techniques , Mutation, Missense , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Skin Diseases, Genetic/genetics , Skin Diseases, Genetic/physiopathology , Transfection
5.
Cell Death Differ ; 9(9): 995-1006, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12181750

ABSTRACT

Caspase-14 is a recent addition to the caspase family of aspartate proteases involved in apoptotic processes. Human caspase-14 appears to be only weakly processed during apoptosis, and it does not cleave classical caspase substrates. Post partum, caspase-14 is prominently expressed by human keratinocytes and reportedly participates in terminal differentiation of complex epithelia. Here we provide evidence challenging the view that caspase-14 expression or processing is linked exclusively to terminal keratinocyte differentiation. We demonstrate that caspase-14 expression extended to multiple cell lines derived from simple epithelia of the breast, prostate, and stomach. In keratinocytes and breast epithelial cells, caspase-14 expression was upregulated in high-density cultures and during forced suspension culture. These effects were primarily due to transcriptional activation as indicated by reporter gene assays using a 2 kb caspase-14 promoter fragment. Importantly, caspase-14 was not cleaved during forced suspension culture of either cell type although this treatment induced caspase-dependent apoptosis (anoikis). Forced expression of caspase-14 in immortalized human keratinocytes had no effect on cell death in forced suspension nor was the transfected caspase-14 processed in this setting. In contrast to postconfluent and forced suspension culture, terminal differentiation of keratinocytes induced in vitro by Ca2+ treatment was not associated with increased caspase-14 expression or promoter activity. Our results indicate that (1) caspase-14 is expressed not only in complex but also simple epithelia; (2) cells derived from complex and simple epithelia upregulate caspase-14 expression in conditions of high cell density or lack of matrix interaction and; (3) in both cell types this phenomenon is due to transcriptional regulation.


Subject(s)
Caspases/genetics , Cell Differentiation/genetics , Epithelial Cells/enzymology , Epithelium/enzymology , Gene Expression Regulation, Enzymologic/genetics , Genes, Regulator/genetics , Antibody Specificity/immunology , Breast/cytology , Breast/enzymology , Breast/growth & development , Caspase 14 , Cell Adhesion/physiology , Cell Compartmentation/physiology , Cell Cycle/physiology , Cells, Cultured , Epidermal Cells , Epidermis/enzymology , Epidermis/growth & development , Epithelial Cells/cytology , Epithelium/growth & development , Extracellular Matrix/enzymology , Humans , Infant, Newborn , Keratinocytes/cytology , Keratinocytes/enzymology , Male , Promoter Regions, Genetic/genetics , Prostate/cytology , Prostate/enzymology , Prostate/growth & development
6.
Matrix Biol ; 20(8): 577-87, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11731274

ABSTRACT

Matrix metalloproteinase-8 (MMP-8) is a neutral metalloproteinase of the fibrillar collagenase family that also includes MMP-1 and MMP-13. In contrast to the other collagenases, MMP-8 has a very limited tissue distribution, thought to be restricted to neutrophils and chondrocytes. In a previous study, we observed MMP-8 expression in human melanoma cells. This observation led us to assess in more detail the expression of MMP-8 in normal and malignant melanocytic cells. We found that MMP-8 was expressed by 11 out of 12 human melanoma cell lines tested and all 10 primary melanomas we examined, but was not expressed by four primary neonatal melanocyte strains. Since melanocytes arise from highly motile neural crest cells, we examined the hypothesis that MMP-8 might be expressed by neural crest cells. RT-PCR analysis of post-implantation mouse embryos indicated the presence of MMP-8 transcripts at E9.5. In situ hybridization and immunohistochemistry of mouse embryos between E9.5-E14.5 demonstrated MMP-8 expression in the surface ectoderm, neural crest cells and chondrocytes. MMP-8 was also detected in neural crest cell migration located in the circumference of the neural tube, branchial arches and the notochord. In addition, MMP-8 expression was observed between the somites, in circumscriptive areas of the developing brain, heart, and eye, and in the interdigital zones of the limbs. In summary, we found MMP-8 to be the first fibrillar collagenase expressed during development. In contrast to its restricted tissue expression post-partum, MMP-8 was present in multiple embryonic tissues, including neural crest cells. The production of MMP-8 by migrating neural crest cells may contribute to their ability to degrade fibrillar collagen matrices while in transit.


Subject(s)
Matrix Metalloproteinase 8/genetics , Matrix Metalloproteinase 8/metabolism , Melanoma/enzymology , Melanoma/genetics , Neural Crest/enzymology , Adult , Animals , Cartilage/enzymology , Embryonic and Fetal Development/genetics , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Humans , Immunohistochemistry , In Situ Hybridization , Melanocytes/enzymology , Mice , Neural Crest/cytology , Neutrophils/enzymology , Tumor Cells, Cultured
7.
Mol Biol Cell ; 12(11): 3328-39, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11694570

ABSTRACT

The Forkhead family of transcription factors participates in the induction of death-related genes. In NMuMG and 4T1 mammary epithelial cells, transforming growth factor beta (TGF beta) induced phosphorylation and cytoplasmic retention of the Forkhead factor FKHRL1, while reducing FHKRL1-dependent transcriptional activity. TGF beta-induced FKHRL1 phosphorylation and nuclear exclusion were inhibited by LY294002, an inhibitor of phosphatidylinositol-3 kinase. A triple mutant of FKHRL1, in which all three Akt phosphorylation sites have been mutated (TM-FKHRL1), did not translocate to the cytoplasm in response to TGF beta. In HaCaT keratinocytes, expression of dominant-negative Akt prevented TGF beta-induced 1) reduction of Forkhead-dependent transcription, 2) FKHRL1 phosphorylation, and 3) nuclear exclusion of FKRHL1. Forced expression of either wild-type (WT) or TM-FKHRL1, but not a FKHRL1 mutant with deletion of the transactivation domain, resulted in NMuMG mammary cell apoptosis. Evidence of nuclear fragmentation colocalized to cells with expression of WT- or TM-FKHRL1. The apoptotic effect of WT-FKHRL1 but not TM-FKHRL1 was prevented by exogenous TGF beta. Serum starvation-induced apoptosis was also inhibited by TGF beta in NMuMG and HaCaT cells. Finally, dominant-negative Akt abrogated the antiapoptotic effect of TGF beta. Taken together, these data suggest that TGF beta may play a role in epithelial cell survival via Akt-dependent regulation of FKHRL1.


Subject(s)
Apoptosis , DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Active Transport, Cell Nucleus , Animals , Cell Division , Cell Nucleus/metabolism , Cell Survival , Epithelial Cells/cytology , Forkhead Box Protein O3 , Forkhead Transcription Factors , Mice , Phosphorylation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Transcription, Genetic
8.
Cancer Res ; 61(19): 7282-90, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11585767

ABSTRACT

Human melanoma cells express two prominent angiogenic factors, e.g., vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF/fibroblast growth factor-2). In this study, we report on the relative contribution of these two factors to in vitro and in vivo growth of a tumorigenic melanoma cell line (WM164) and nontumorigenic, immortalized melanocytes (FM516SV). Overexpression of either cytokine significantly boosted tumorigenicity of WM164 cells in immunodeficient SCID mice. Attempting to overexpress bFGF antisense sequences produced no viable clones confirming earlier reports that autocrine bFGF is obligatory to melanoma cell survival and growth. By contrast, down-regulation of endogenous VEGF production did not affect growth of WM164 cells in vitro. In vivo expansion of WM164 cells expressing VEGF antisense was delayed but not abrogated. Forced expression of either bFGF or VEGF in immortalized but nontumorigenic melanocytes did not induce sustained tumor growth in vivo highlighting that neither of the two factors is sufficient for induction of tumorigenicity in this model system. Overexpression of either cytokine in WM164 cells led to the development of atypical large vessels but not to an increase in microvessel density. Taken together our results confirm an essential autocrine role of bFGF in human melanoma and indicate a beneficial but nonessential role of VEGF in the tumorigenic phenotype of human melanoma cells.


Subject(s)
Endothelial Growth Factors/physiology , Fibroblast Growth Factor 2/physiology , Lymphokines/physiology , Melanocytes/physiology , Melanoma/blood supply , Neovascularization, Pathologic/metabolism , Animals , Cell Line , Cell Transformation, Neoplastic , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/genetics , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiology , Fibroblast Growth Factor 2/biosynthesis , Fibroblast Growth Factor 2/genetics , Gene Expression , Humans , Immunohistochemistry , Lymphokines/biosynthesis , Lymphokines/genetics , Melanocytes/cytology , Melanocytes/metabolism , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, SCID , Neovascularization, Pathologic/genetics , Transfection , Transgenes , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
9.
Gene ; 273(1): 29-39, 2001 Jul 25.
Article in English | MEDLINE | ID: mdl-11483358

ABSTRACT

The human metastasis-associated gene (MTA1) is overexpressed in cell lines and tissues representing metastatic tumors. Here we report cloning of the mouse Mta1 as well as a novel structurally related mouse gene, Mta3. The mouse Mta1 protein shares 94 and 59% homology to the human MTA1 and mouse Mta3 proteins, respectively. Northern blotting analysis using an Mta1 cDNA probe revealed a prevalent 3 kb hybridization signal in all mouse tissues except the skeletal muscle while a smaller approximately 1.0 kb mRNA product was also detected in the heart. Mta3 transcripts (approximately 2 kb) were detected in most tissues with an additional approximately 6.2 kb signal detected in the brain. In vitro transcription/translation of the full-length Mta1 and Mta3 cDNAs generated products of the expected molecular masses, i.e. 80 and 60 kDa, respectively. To assess subcellular localization, green fluorescence protein (GFP)-tagged expression constructs of Mta1 and Mta3 and various deletion constructs of GFP-Mta1 were transiently expressed in Balb/MK keratinocytes. GFP-Mta1 was found exclusively in the nucleus while GFP-Mta3 was present in both the nucleus and cytoplasm. Compared to Mta3, the carboxy terminal end of Mta1 contains an additional nuclear localization signal (NLS) and a proline-rich Src homology 3 (SH3) ligand. The results of transient expression experiments of various Mta1 fragments containing these domains in different combinations indicated that nuclear localization of Mta1 depended on the presence of at least one NLS and one SH3 binding site. These SH3 ligands appeared to be functional as they facilitated interaction with the adaptor protein, Grb2, and the Src-family tyrosine kinase, Fyn.


Subject(s)
Histone Deacetylases , Neoplasm Proteins/metabolism , Proteins/metabolism , Repressor Proteins , Amino Acid Sequence , Animals , Cell Nucleus/metabolism , Cloning, Molecular , Cytoplasm/metabolism , Humans , Keratinocytes/metabolism , Mice , Molecular Sequence Data , Nuclear Localization Signals , Proteins/genetics , Sequence Homology , Trans-Activators , Transfection , src Homology Domains
10.
J Invest Dermatol ; 116(6): 860-6, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11407972

ABSTRACT

Previous work has shown that activation of the epidermal growth factor receptor by endogenous or exogenous signals markedly enhances survival of cultured keratinocytes upon cellular stress such as passaging. This is due, in part, to epidermal-growth-factor-receptor-dependent expression of Bcl-x(L), an antiapoptotic Bcl-2 homolog. In this study we tested whether epidermal-growth-factor-receptor-dependent signal transduction and attendant Bcl-x(L) expression affected survival of human keratinocytes upon exposure to a frequently encountered apoptotic stimulus, radiation with ultraviolet B. We describe that blocking epidermal-growth-factor-receptor-dependent signal transduction sensitized normal keratinocytes to undergo apoptosis upon ultraviolet B radiation with solar light characteristics. Forced expression of Bcl-x(L) partially but significantly inhibited ultraviolet-B-induced apoptosis of immortalized keratinocytes (HaCaT). Bcl-x(L) overexpression afforded no protection to HaCaT cells against apoptosis induced by binding of an agonist antibody to the death receptor CD95, however. CD95 activation has previously been shown to functionally contribute to apoptosis in ultraviolet-irradiated keratinocytes. These results indicate that epidermal growth factor receptor activation and attendant Bcl-x(L) expression provided a physiologically relevant protective pathway of keratinocytes against ultraviolet-induced but not CD95-dependent apoptosis.


Subject(s)
Apoptosis/radiation effects , ErbB Receptors/physiology , Keratinocytes/radiation effects , Proto-Oncogene Proteins c-bcl-2/physiology , Radiation Protection , Ultraviolet Rays , fas Receptor/physiology , Cells, Cultured , Humans , Keratinocytes/physiology , bcl-X Protein
11.
Mol Biol Cell ; 12(5): 1519-27, 2001 May.
Article in English | MEDLINE | ID: mdl-11359940

ABSTRACT

Normal epithelial cells undergo apoptosis when they are denied contact with the extracellular matrix, in a process termed "anoikis." Conversely, malignant epithelial cells typically acquire anchorage independence, i.e., the capacity to survive and grow in the absence of matrix interaction. Here we asked the question whether anoikis is affected by signaling through the EGF receptor (EGFR). We focused on the EGFR because EGFR signaling is frequently deregulated in malignant epithelial cells. We demonstrate that EGFR activation markedly alleviated the requirement of matrix engagement for survival of primary and immortalized human keratinocytes in suspension culture. Protection of epithelial cells through EGFR activation against anoikis was associated with and required sustained MAPK phosphorylation during the early phase of suspension culture. Interestingly, high levels of MAPK phosphorylation were not only required for EGFR-mediated protection against anoikis but also occurred as a consequence of caspase activation at later stages of suspension culture. These results demonstrate that EGFR activation contributes to anchorage-independent epithelial cell survival and identify MAPK activation as an important mechanism in this process.


Subject(s)
ErbB Receptors/metabolism , Extracellular Matrix/physiology , Keratinocytes/metabolism , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Caspase Inhibitors , Caspases/metabolism , Cell Survival , Cells, Cultured , Chromones/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/pharmacology , Flavonoids/pharmacology , Humans , In Situ Nick-End Labeling , Keratinocytes/cytology , Keratinocytes/drug effects , Mitogen-Activated Protein Kinase Kinases/metabolism , Morpholines/pharmacology , Oligopeptides/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Tyrosine Phosphatases/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , Quinazolines , Tyrphostins/pharmacology , bcl-X Protein
12.
Cancer Res ; 61(5): 2200-6, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11280787

ABSTRACT

Multiple genetic aberrations contribute to the development of biologically aggressive, clinically malignant colorectal carcinomas (CRCs). Some of these have been linked to inappropriate signaling through the tyrosine kinase moieties of growth factor receptors. We have described previously (G. Bellone et al., J. Cell. Physiol., 172: 1-11, 1997) that human CRCs overexpress both the receptor tyrosine kinase c-kit and its ligand, stem cell factor (SCF), relative to normal mucosa cells, thus establishing an autocrine c-kit-mediated loop. In addition, we noted that exogenous SCF contributes to anchorage-independent growth of HT-29 colon carcinoma cells in semisolid medium. Here, we investigated possible roles of the c-kit/SCF autocrine/paracrine system in survival and invasive capacity of DLD-1 colon carcinoma cells. We report that SCF was required for migration and invasion of DLD-1 cells through reconstituted basement membranes (Matrigel) and up-regulated gelatinase (matrix metalloproteinase-9) activity in DLD-1 cells. Furthermore, we describe that SCF supported survival of DLD-1 cells in growth factor-deprived conditions. These results suggest multiple roles of c-kit activation in support of the malignant phenotype of DLD-1 cells related to growth, survival, migration, and invasive potential.


Subject(s)
Apoptosis/physiology , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Proto-Oncogene Proteins c-kit/physiology , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Division/drug effects , Cell Division/physiology , Cell Movement/drug effects , Cell Movement/physiology , Colonic Neoplasms/metabolism , Down-Regulation/drug effects , Down-Regulation/physiology , Enzyme Activation , Humans , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/metabolism , Neoplasm Invasiveness , Oligonucleotides, Antisense/pharmacology , Phenotype , Proto-Oncogene Proteins c-kit/metabolism , Stem Cell Factor/metabolism , Stem Cell Factor/pharmacology , Stem Cell Factor/physiology , Tumor Cells, Cultured
13.
Eur J Cancer ; 37(2): 224-33, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11166150

ABSTRACT

Transforming growth factor (TGF)-beta is a protein family which affects multiple cellular functions including survival, proliferation, differentiation and adhesion. Among the three known isoforms, TGF-beta1 is commonly overexpressed in solid malignancies. Recent studies in knock-out mice demonstrated non-redundant roles of different TGF-beta isoforms in development. The present study was performed to assess tumour-associated expression of the three TGF-beta isoforms in colon carcinoma. We report that colon carcinoma progression is associated with gradual and significant increases in expression of TGF-beta1 and TGF-beta2 mRNA and proteins. By contrast, TGF-beta3 expression was detected in normal colonic mucosa and, at slightly higher levels, in tumour tissues. In addition, plasma levels of both TGF-beta1 and TGF-beta2 were significantly higher in cancer patients when compared with unaffected individuals. Taken together, our results indicate distinct expression patterns of the three TGF-beta isoforms in colon carcinoma cells and possible systemic effects of TGF-beta1 and TGF-beta2 in tumour patients.


Subject(s)
Carcinoma in Situ/diagnosis , Colonic Neoplasms/diagnosis , Neoplasm Proteins/metabolism , Transforming Growth Factor beta/metabolism , Adult , Aged , Aged, 80 and over , Disease Progression , Female , Humans , Immunohistochemistry , Lymphatic Metastasis , Male , Middle Aged , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta1 , Transforming Growth Factor beta2 , Transforming Growth Factor beta3
14.
J Biol Chem ; 276(9): 6320-6, 2001 Mar 02.
Article in English | MEDLINE | ID: mdl-11098053

ABSTRACT

Previous work has shown that the epidermal growth factor receptor (EGFR) tyrosine kinase moiety provides protection to normal human keratinocytes against apoptosis. This protection is, at least in part, due to EGFR-dependent expression of the antiapoptotic Bcl-2 family member, Bcl-x(L). Here we focused on intracellular signaling pathways relevant to keratinocyte survival and/or Bcl-x(L) expression. By using pharmacological inhibitors and dominant negative expression constructs, we observed that phosphatidylinositol 3-kinase/AKT and phospholipase C gamma/protein kinase C alpha activation were required for keratinocyte survival independently of EGFR activation or Bcl-x(L) expression. By contrast, MEK activity required EGFR activation and, as shown by use of the MEK inhibitor PD98059 and a dominant negative MEK construct, was necessary for Bcl-x(L) expression and survival. Consistent with an earlier study, blocking SRC kinase activities similarly led to down-regulation of Bcl-x(L) protein expression and impaired keratinocyte survival. In conclusion, our results demonstrate that EGFR-dependent MEK activity contributes to both Bcl-x(L) expression and survival of normal keratinocytes. Other signaling pathways (i.e. phosphatidylinositol 3-kinase/AKT and phospholipase C gamma/protein kinase C alpha) are obligatory to keratinocyte survival but not to Bcl-x(L) expression, and control of these pathways by EGFR activation is not rate-limiting to normal keratinocyte survival.


Subject(s)
ErbB Receptors/physiology , Keratinocytes/physiology , Mitogen-Activated Protein Kinase Kinases/physiology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Cell Line , Cell Survival , DNA-Binding Proteins/physiology , Humans , MAP Kinase Kinase 1 , Phosphatidylinositol 3-Kinases/physiology , STAT3 Transcription Factor , Signal Transduction , Trans-Activators/physiology , Type C Phospholipases/physiology , bcl-X Protein
15.
Eur J Dermatol ; 10(7): 505-10, 2000.
Article in English | MEDLINE | ID: mdl-11056418

ABSTRACT

Epidermal keratinocytes express both the epidermal growth factor receptor (EGFR) and several of its ligands, establishing the constitutive elements of an autocrine loop in this cell type. Activation of the EGFR provides signals essential to several aspects of normal keratinocyte biology including cell cycle progression, differentiation, cell movement and cellular survival. It may be argued that enhanced keratinocyte survival via EGFR activation is the most important function as it limits the manifestation of other phenotypes. The frequent deregulation of EGFR expression and activation in benign and malignant hyperproliferative skin diseases motivates the investigation of EGFR-dependent intracellular pathways which contribute to the varied EGFR-dependent phenotypes.


Subject(s)
Cell Movement/physiology , Epidermis/physiology , ErbB Receptors/physiology , Keratinocytes/physiology , Cell Division/physiology , Cell Survival , Cells, Cultured , Epidermal Cells , ErbB Receptors/analysis , Humans , Sensitivity and Specificity , Signal Transduction
16.
J Invest Dermatol ; 115(1): 118-23, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10886518

ABSTRACT

Expression patterns of the angiogenic placenta growth factor and its receptor neuropilin-1 were assessed in normal human melanocytes, SV40T-transformed melanocytes, and melanoma cells derived from primary and metastatic lesions. As determined by reverse transcription-polymerase chain reaction all primary and metastatic melanoma cell lines tested and SV40T-transformed melanocytes coexpressed two placenta growth factor splice variants (placenta growth factor-1 and -2) as well as neuropilin-1 mRNA. Placenta growth factor protein was detected in conditioned media derived from five of eight melanomas and from SV40T-transformed melanocytes. In contrast to melanoma cells, normal melanocytes did not express placenta growth factor mRNA at detectable levels and did not secrete placenta growth factor protein. By contrast, neuropilin-1 transcripts were detected in some of the normal melanocytes. Secretion of placenta growth factor by melanoma cells appeared to be constitutive because it was not affected by the addition of exogenous growth factors including insulin, epidermal growth factor, or basic fibroblast growth factor to culture media. Although melanoma cells expressed both, neuropilin-1 and flt-1, exogenous homodimeric placenta growth factor had no effect on melanoma cell growth. Similarly, placenta growth factor did not induce urokinase-type plasminogen activator production in these cells. These findings demonstrate that melanoma progression is accompanied by deregulated, constitutive placenta growth factor expression. Placenta growth factor, however, serves no apparent autocrine role in melanoma proliferation. Further studies are needed to define the relative contribution of placenta growth factor to the angiogenic properties of human melanomas.


Subject(s)
Angiogenesis Inducing Agents/biosynthesis , Melanocytes/cytology , Melanocytes/metabolism , Pregnancy Proteins/biosynthesis , Cell Line , Cell Transformation, Neoplastic/metabolism , Growth Substances/biosynthesis , Humans , Melanoma/chemistry , Melanoma/pathology , Placenta/chemistry , Placenta Growth Factor
17.
Clin Cancer Res ; 6(6): 2448-55, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10873098

ABSTRACT

We report here that the progression of pancreatic carcinomas in tumor patients is associated with increased serum levels of both the soluble forms of CD95 ligand (CD95L/FasL) and its receptor, CD95 (Fas). Shedding of proteolytically processed soluble CD95L was also observed in pancreatic carcinoma cells in vitro, thus identifying one possible source of CD95L in patients' sera. Because the secreted forms of both CD95 and CD95L have been implicated previously in protection of cells from CD95-mediated cell death, we assessed the effect of soluble CD95L in supernatants of pancreatic carcinoma cells on viability of Jurkat T lymphocytes. We describe that (a) supernatants derived from cultured pancreatic carcinoma cells caused apoptosis of Jurkat cells; (b) soluble tumor-derived CD95L contributed significantly to this effect; and (c) in comparison to Jurkat cells, pancreatic carcinoma cells themselves revealed increased resistance to apoptosis induction by autocrine soluble CD95L. These results are consistent with the notion that in the microenvironment of pancreatic tumors, tumor-derived shed CD95L exerts paracrine pro-apoptotic effects. In addition, because it is released at high levels into the bloodstream, soluble CD95L may have systemic effects in tumor patients that reach beyond the microenvironment of the tumor site.


Subject(s)
Apoptosis , Carcinoma/metabolism , Membrane Glycoproteins/biosynthesis , Pancreatic Neoplasms/metabolism , fas Receptor/biosynthesis , Adult , Aged , Carcinoma/blood , Cell Separation , Coculture Techniques , Culture Media, Conditioned , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Fas Ligand Protein , Female , Flow Cytometry , Fluorescent Antibody Technique , Green Fluorescent Proteins , Humans , Immunoblotting , Immunohistochemistry , Jurkat Cells , Luminescent Proteins/metabolism , Male , Membrane Glycoproteins/blood , Middle Aged , Pancreatic Neoplasms/blood , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Tumor Cells, Cultured , fas Receptor/blood
18.
Cell Growth Differ ; 10(11): 721-8, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10593648

ABSTRACT

The extracellular matrix-associated glycoprotein secreted protein acidic and rich in cysteine (SPARC) has been implicated in the control of cell proliferation during tissue remodeling, wound healing, and malignant development. Here, we describe a novel mechanism through which SPARC influences cell cycle progression in embryonic fibroblasts derived from Sparc-nullizygous (-/-) mice. SPARC-deficient cells were indistinguishable from wild-type cells in their ability to initiate DNA synthesis after treatment with either fetal bovine serum or platelet-derived growth factor. In contrast, Sparc -/- cells responded poorly to activation of the insulin-like growth factor receptor (IGFI-R) by insulin. This defect was traced to reduced expression of the IGFI-R in Sparc -/- cells. Consistent with impaired cell cycle progression through S-phase, insulin-stimulated Sparc -/- cells also revealed reduced expression of two key regulators of S phase progression (cyclin A and thymidine kinase), whereas expression of the G1 phase progression regulators cmyc or cyclin D1 was unaffected. An examination of the status of retinoblastoma family pocket proteins in Sparc -/- cells revealed a selective and dramatic reduction in levels of the retinoblastoma-related protein p107. Exogenous platelet-derived growth factor restored expression of the IGFI-R and IGFI-R dependent DNA synthesis as well as induction of cyclin A, thymidine kinase, and p107 in insulin-stimulated Sparc -/- cells. These results suggest that SPARC-dependent matrix to cell interactions contribute to the regulation of p107 and cyclin A through IGFI-R dependent pathway(s).


Subject(s)
Cyclin A/genetics , Nuclear Proteins/genetics , Osteonectin/genetics , Receptor, IGF Type 1/genetics , Animals , Cell Division/drug effects , Cell Division/genetics , Cells, Cultured , Cysteine , Extracellular Matrix/chemistry , Extracellular Matrix/enzymology , Extracellular Matrix/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/enzymology , Gene Expression Regulation/physiology , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteonectin/metabolism , Platelet-Derived Growth Factor/pharmacology , Retinoblastoma-Like Protein p107 , S Phase/physiology , Specific Pathogen-Free Organisms , Thymidine Kinase/genetics , Transcriptional Activation/physiology
19.
J Cancer Res Clin Oncol ; 125(11): 621-9, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10541969

ABSTRACT

The expression patterns of vascular endothelial growth factor (VEGF) and its two receptors, flt-1 and KDR, were assessed in normal human melanocytes, transformed melanocytes expressing the simian virus 40 Tgene (SV40T), and melanoma cells derived from primary and metastatic lesions. Constitutive expression of VEGF, flt-1, and KDR mRNA and proteins was observed in the majority of primary and metastatic melanoma cell lines, and in SV40T-transformed melanocytes. VEGF expression in melanoma cell lines was further enhanced by exogenous growth factors including insulin and fetal calf serum. By contrast, neonatal melanocytes did not express VEGF or VEGF receptors and VEGF expression could not be induced by exogenous growth factors. Exogenous VEGF had no significant effects on melanoma cell proliferation or on production of a transcriptional target for VEGF, urokinase-type plasminogen activator. Down-regulation of VEGF expression in the metastatic melanoma cell line WM164 through transfection of a VEGF antisense construct similarly did not affect proliferation of the transfected cells in the presence or absence of exogenous VEGF. In summary, coexpression of VEGF and its receptors is a tumor-associated phenomenon in melanoma development. However VEGF production does not support autocrine proliferation of the melanoma cell lines tested.


Subject(s)
Endothelial Growth Factors/biosynthesis , Lymphokines/biosynthesis , Melanoma/metabolism , Proto-Oncogene Proteins/biosynthesis , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptors, Growth Factor/biosynthesis , Antibody Specificity , Cell Division/drug effects , Cells, Cultured , Culture Media, Conditioned/metabolism , DNA, Antisense/pharmacology , Endothelial Growth Factors/genetics , Endothelial Growth Factors/metabolism , Endothelial Growth Factors/pharmacology , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Lymphokines/genetics , Lymphokines/metabolism , Lymphokines/pharmacology , RNA, Messenger/biosynthesis , Receptors, Vascular Endothelial Growth Factor , Reverse Transcriptase Polymerase Chain Reaction , Urokinase-Type Plasminogen Activator/metabolism , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1 , Vascular Endothelial Growth Factors
20.
Am J Pathol ; 155(2): 537-47, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10433946

ABSTRACT

In this study, we report coexpression of transforming growth factor-beta (TGF-beta) and interleukin-10 (IL-10) in pancreatic carcinoma tissue associated with significantly elevated levels of both cytokines in the sera of pancreatic carcinoma patients. Using conditioned media (CM) of pancreatic carcinoma cells, we further demonstrate that tumor cell-derived TGF-beta and IL-10 inhibited in an additive fashion both proliferation and the development of Th1-like responses in peripheral blood mononuclear cell (PBMC) preparations derived from normal donors. The antiproliferative and Th1-suppressive activities contained in CM of pancreatic carcinoma cells were due primarily to IL-10 and/or TGF-beta, as shown by the capacity of cytokine-specific neutralizing antibodies to reverse these effects. Finally, as compared to normal controls, PBMC derived from pancreatic carcinoma patients displayed a Th2-like cytokine expression pattern upon activation with either anti-CD3 antibody or Staphylococcus aureus strain Cowan I. Taken together, these results suggest that aberrant production of TGF-beta and IL-10 in pancreatic tumor patients skews T-cell cytokine production patterns in favor of a Th2 immunophenotype.


Subject(s)
Adenocarcinoma/metabolism , Interleukin-10/metabolism , Pancreatic Neoplasms/metabolism , T-Lymphocytes/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Transforming Growth Factor beta/metabolism , Adenocarcinoma/immunology , Aged , Antibodies, Monoclonal , Culture Media, Conditioned/pharmacology , Dose-Response Relationship, Drug , Female , Humans , Immunophenotyping , Interferon-gamma/antagonists & inhibitors , Interleukin-10/biosynthesis , Interleukin-10/blood , Interleukin-12/antagonists & inhibitors , Killer Cells, Lymphokine-Activated/immunology , Male , Middle Aged , Pancreatic Ducts/pathology , Pancreatic Neoplasms/immunology , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/blood , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...