Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Cell Commun Signal ; 16(1): 36, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29966518

ABSTRACT

BACKGROUND: Chemotherapeutic efficacy can be improved by targeting the structure and function of the extracellular matrix (ECM) in the carcinomal stroma. This can be accomplished by e.g. inhibiting TGF-ß1 and -ß3 or treating with Imatinib, which results in scarcer collagen fibril structure in xenografted human KAT-4/HT29 (KAT-4) colon adenocarcinoma. METHODS: The potential role of αVß6 integrin-mediated activation of latent TGF-ß was studied in cultured KAT-4 and Capan-2 human ductal pancreatic carcinoma cells as well as in xenograft carcinoma generated by these cells. The monoclonal αVß6 integrin-specific monoclonal antibody 3G9 was used to inhibit the αVß6 integrin activity. RESULTS: Both KAT-4 and Capan-2 cells expressed the αVß6 integrin but only KAT-4 cells could utilize this integrin to activate latent TGF-ß in vitro. Only when Capan-2 cells were co-cultured with human F99 fibroblasts was the integrin activation mechanism triggered, suggesting a more complex, fibroblast-dependent, activation pathway. In nude mice, a 10-day treatment with 3G9 reduced collagen fibril thickness and interstitial fluid pressure in KAT-4 but not in the more desmoplastic Capan-2 tumors that, to achieve a similar effect, required a prolonged 3G9 treatment. In contrast, a 10-day direct inhibition of TGF-ß1 and -ß3 reduced collagen fibril thickness in both tumor models. CONCLUSION: Our data demonstrate that the αVß6-directed activation of latent TGF-ß plays a pivotal role in modulating the stromal collagen network in carcinoma, but that the sensitivity to αVß6 inhibition depends on the simultaneous presence of alternative paths for latent TGF-ß activation and the extent of desmoplasia.


Subject(s)
Antigens, Neoplasm/immunology , Collagen/chemistry , Integrins/immunology , Animals , Antibodies, Monoclonal/immunology , Antigens, Neoplasm/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic , Collagen/metabolism , Extracellular Fluid/metabolism , Female , Gene Expression Profiling , Humans , Integrins/metabolism , Mice , Pressure , Transforming Growth Factor beta/metabolism
2.
PLoS One ; 7(3): e34082, 2012.
Article in English | MEDLINE | ID: mdl-22479530

ABSTRACT

Stroma properties affect carcinoma physiology and direct malignant cell development. Here we present data showing that α(V)ß(3) expressed by stromal cells is involved in the control of interstitial fluid pressure (IFP), extracellular volume (ECV) and collagen scaffold architecture in experimental murine carcinoma. IFP was elevated and ECV lowered in syngeneic CT26 colon and LM3 mammary carcinomas grown in integrin ß(3)-deficient compared to wild-type BALB/c mice. Integrin ß(3)-deficiency had no effect on carcinoma growth rate or on vascular morphology and function. Analyses by electron microscopy of carcinomas from integrin ß(3)-deficient mice revealed a coarser and denser collagen network compared to carcinomas in wild-type littermates. Collagen fibers were built from heterogeneous and thicker collagen fibrils in carcinomas from integrin ß(3)-deficient mice. The fibrotic extracellular matrix (ECM) did not correlate with increased macrophage infiltration in integrin ß(3)-deficient mice bearing CT26 tumors, indicating that the fibrotic phenotype was not mediated by increased inflammation. In conclusion, we report that integrin ß(3)-deficiency in tumor stroma led to an elevated IFP and lowered ECV that correlated with a more fibrotic ECM, underlining the role of the collagen network for carcinoma physiology.


Subject(s)
Carcinoma/metabolism , Fibrosis/pathology , Gene Expression Regulation, Neoplastic , Integrin beta3/genetics , Animals , Cell Line, Tumor , Collagen/metabolism , Extracellular Fluid , Female , Hydroxyproline/metabolism , Integrin alphaVbeta3/metabolism , Male , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Phenotype , Pressure
3.
Clin Cancer Res ; 17(23): 7248-64, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21868764

ABSTRACT

PURPOSE: Human cell lines are useful for studying cancer biology and preclinically modeling cancer therapy, but can be misidentified and cross-contamination is unfortunately common. The purpose of this study was to develop a panel of validated head and neck cell lines representing the spectrum of tissue sites and histologies that could be used for studying the molecular, genetic, and phenotypic diversity of head and neck cancer. METHODS: A panel of 122 clinically and phenotypically diverse head and neck cell lines from head and neck squamous cell carcinoma, thyroid cancer, cutaneous squamous cell carcinoma, adenoid cystic carcinoma, oral leukoplakia, immortalized primary keratinocytes, and normal epithelium was assembled from the collections of several individuals and institutions. Authenticity was verified by carrying out short tandem repeat analysis. Human papillomavirus (HPV) status and cell morphology were also determined. RESULTS: Eighty-five of the 122 cell lines had unique genetic profiles. HPV-16 DNA was detected in 2 cell lines. These 85 cell lines included cell lines from the major head and neck primary tumor sites, and close examination shows a wide range of in vitro phenotypes. CONCLUSIONS: This panel of 85 genomically validated head and neck cell lines represents a valuable resource for the head and neck cancer research community that can help advance understanding of the disease by providing a standard reference for cell lines that can be used for biological as well as preclinical studies.


Subject(s)
Carcinoma, Squamous Cell , Cell Line, Tumor , Head and Neck Neoplasms , Alphapapillomavirus/genetics , Alphapapillomavirus/isolation & purification , Carcinoma, Adenoid Cystic/genetics , Carcinoma, Squamous Cell/genetics , Cell Culture Techniques , DNA, Viral/analysis , Head and Neck Neoplasms/genetics , Humans , Keratinocytes/cytology , Leukoplakia, Oral/genetics , Microsatellite Repeats , Thyroid Neoplasms/genetics
4.
Biol Chem ; 391(8): 923-35, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20536394

ABSTRACT

The cysteine peptidase cathepsin B is important in thyroid physiology by being involved in thyroid prohormone processing initiated in the follicular lumen and completed in endo-lysosomal compartments. However, cathepsin B has also been localized to the extrafollicular space and is therefore suggested to promote invasiveness and metastasis in thyroid carcinomas through, e.g., ECM degradation. In this study, immunofluorescence and biochemical data from subcellular fractionation revealed that cathepsin B, in its single- and two-chain forms, is localized to endo-lysosomes in the papillary thyroid carcinoma cell line KTC-1 and in the anaplastic thyroid carcinoma cell lines HTh7 and HTh74. This distribution is not affected by thyroid stimulating hormone (TSH) incubation of HTh74, the only cell line that expresses a functional TSH-receptor. Immunofluorescence data disclosed an additional nuclear localization of cathepsin B immunoreactivity. This was supported by biochemical data showing a proteolytically active variant slightly smaller than the cathepsin B proform in nuclear fractions. We also demonstrate that immunoreactions specific for cathepsin V, but not cathepsin L, are localized to the nucleus in HTh74 in peri-nucleolar patterns. As deduced from co-localization studies and in vitro degradation assays, we suggest that nuclear variants of cathepsins are involved in the development of thyroid malignancies through modification of DNA-associated proteins.


Subject(s)
Carcinoma/enzymology , Cathepsin B/metabolism , Cell Nucleus/enzymology , Genetic Variation , Thyroid Neoplasms/enzymology , Carcinoma/metabolism , Carcinoma/pathology , Cathepsin B/chemistry , Cathepsins/chemistry , Cathepsins/metabolism , Cell Fractionation , Cell Line, Tumor , Cell Nucleus/pathology , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/metabolism , Humans , Isoenzymes/chemistry , Isoenzymes/metabolism , Lysosomes/enzymology , Microscopy, Confocal , Microscopy, Fluorescence , Microscopy, Phase-Contrast , Molecular Weight , Nuclear Proteins/metabolism , Protein Subunits/metabolism , RNA, Messenger/metabolism , Receptors, Thyrotropin/genetics , Receptors, Thyrotropin/metabolism , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Thyrotropin/metabolism
5.
PLoS One ; 4(9): e7133, 2009 Sep 22.
Article in English | MEDLINE | ID: mdl-19771160

ABSTRACT

BACKGROUND: BMPs are currently receiving attention for their role in tumorigenesis and tumor progression. Currently, most BMP expression studies are performed on carcinomas, and not much is known about the situation in sarcomas. METHODOLOGY/PRINCIPAL FINDINGS: We have investigated the BMP expression profiles and Smad activation in clones from different spontaneous canine mammary tumors. Spindle cell tumor and osteosarcoma clones expressed high levels of BMPs, in particular BMP-2, -4 and -6. Clones from a scirrhous carcinoma expressed much lower BMP levels. The various clones formed different tumor types in nude mice but only clones that expressed high levels of BMP-6 gave bone formation. Phosphorylated Smad-1/5, located in the nucleus, was detected in tumors derived from clones expressing high levels of BMPs, indicating an active BMP signaling pathway and BMP-2 stimulation of mammary tumor cell clones in vitro resulted in activation of the Smad-1/5 pathway. In contrast BMP-2 stimulation did not induce phosphorylation of the non-Smad pathway p38 MAPK. Interestingly, an increased level of the BMP-antagonist chordin-like 1 was detected after BMP stimulation of non-bone forming clones. CONCLUSIONS/SIGNIFICANCE: We conclude that the specific BMP expression repertoire differs substantially between different types of mammary tumors and that BMP-6 expression most probably has a biological role in bone formation of canine mammary tumors.


Subject(s)
Bone Morphogenetic Proteins/biosynthesis , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Smad Proteins/metabolism , Animals , Cell Line, Tumor , Dogs , Eye Proteins/metabolism , Female , Mammary Neoplasms, Animal , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Neoplasms, Experimental/metabolism , Nerve Tissue Proteins/metabolism , Phenotype , Transforming Growth Factor beta/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
6.
Breast Cancer Res Treat ; 118(2): 333-43, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19048371

ABSTRACT

The global gene expression in three types of canine mammary tumors: carcinoma, fibrosarcoma and osteosarcoma were investigated by Affymetrix gene array technology. Unsupervised clustering analysis revealed a close clustering of the respective tumor types, with fibrosarcomas clustering close to the osteosarcomas and the carcinomas clustering closer to non-malignant mammary tissues (NMTs). A number of epithelial markers were expressed in both carcinomas and NMTs, whereas the sarcomas expressed genes related to mesenchymal differentiation. A comparison of the gene expression profile of the sarcomas versus carcinoma/NMTs revealed that the sarcomas, in particular the osteosarcomas, showed a striking upregulation of a panel of homeobox genes previously linked to craniofacial bone formation. In line with this finding, osteosarcomas showed an upregulation of bone morphogenetic proteins (BMPs) and of genes associated with retinoic acid signaling. Increased homeobox gene expression in sarcomas was also confirmed at the protein level by immunohistochemical analysis of tumor tissue, and in an osteosarcoma cell line after stimulation by BMP-2. These findings suggest that the development of mammary sarcomas specifically involves triggering of a set of homeobox genes related to neural crest and craniofacial bone development.


Subject(s)
Gene Expression Profiling , Genes, Homeobox/genetics , Mammary Neoplasms, Animal/genetics , Sarcoma/genetics , Animals , Cluster Analysis , Dog Diseases , Dogs , Facial Bones , Female , Fluorescent Antibody Technique , Gene Expression , Immunohistochemistry , Oligonucleotide Array Sequence Analysis , Skull
7.
Proc Natl Acad Sci U S A ; 104(35): 13966-71, 2007 Aug 28.
Article in English | MEDLINE | ID: mdl-17715296

ABSTRACT

Research on the biology of the tumor stroma has the potential to lead to development of more effective treatment regimes enhancing the efficacy of drug-based treatment of solid malignancies. Tumor stroma is characterized by distorted blood vessels and activated connective tissue cells producing a collagen-rich matrix, which is accompanied by elevated interstitial fluid pressure (IFP), indicating a transport barrier between tumor tissue and blood. Here, we show that the collagen-binding proteoglycan fibromodulin controls stroma structure and fluid balance in experimental carcinoma. Gene ablation or inhibition of expression by anti-inflammatory agents showed that fibromodulin promoted the formation of a dense stroma and an elevated IFP. Fibromodulin-deficiency did not affect vasculature but increased the extracellular fluid volume and lowered IFP. Our data suggest that fibromodulin controls stroma matrix structure that in turn modulates fluid convection inside and out of the stroma. This finding is particularly important in relation to the demonstration that targeted modulations of the fluid balance in carcinoma can increase the response to cancer therapeutic agents.


Subject(s)
Collagen/metabolism , Extracellular Matrix Proteins/metabolism , Proteoglycans/metabolism , Animals , Carcinoma/blood supply , Carcinoma/metabolism , Carcinoma/pathology , Extracellular Fluid/physiology , Extracellular Matrix/pathology , Extracellular Matrix/ultrastructure , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/genetics , Fibromodulin , Mice , Mice, Inbred C57BL , Mice, Knockout , Proteoglycans/deficiency , Proteoglycans/genetics , Thyroid Neoplasms/blood supply , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology
8.
Thyroid ; 17(4): 289-301, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17465858

ABSTRACT

In this study we present two novel anaplastic thyroid carcinoma (ATC) lines (HTh 104 and HTh 112) and further characterize six frequently used ATC lines (HTh 7, HTh 74, HTh 83, C 643, KAT-4, and SW 1736). Three of the lines carried a heterozygous BRAF mutation V600E, which is in line with reports of BRAF mutations in primary ATC and papillary thyroid cancer. Several nonrandom breakpoints were identified by spectral karyotyping (SKY) and G-banding in these lines including the novel 1p36 and 17q24-25 as well as 3p21-22 and 15q26 that are also implicated in well-differentiated thyroid cancers. Comparative genomic hybridization showed frequent gain of 20q, including the UBCH10 gene in 20q13.12, which was further confirmed by array-comparative genomic hybridization and fluorescence in situ hybridization analyses. Our results concur with previous studies in both primary tumors and cell lines, indicating that gain of chromosome 20 is important in the pathogenesis of ATC and/or progression of differentiated thyroid cancers to ATC.


Subject(s)
Carcinoma/genetics , Chromosome Aberrations/classification , Chromosomes, Human/genetics , Proto-Oncogene Proteins B-raf/genetics , Thyroid Neoplasms/genetics , Aged , Aged, 80 and over , Carcinoma/pathology , Cell Line, Tumor , Female , Gene Amplification , Gene Expression Regulation, Neoplastic , Humans , Male , Microarray Analysis , Nucleic Acid Hybridization , PTEN Phosphohydrolase/genetics , Spectral Karyotyping , Thyroid Neoplasms/pathology , Ubiquitin-Conjugating Enzymes/genetics
9.
Eur J Nucl Med Mol Imaging ; 34(9): 1376-87, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17277931

ABSTRACT

PURPOSE: We tested the suitability of the chimeric monoclonal anti-human CD44 splice version 6 antibody (cMAb U36) for targeting and visualising human anaplastic thyroid carcinoma with PET. We also performed experiments aimed at elucidating the relation between tumour interstitial fluid pressure (TIFP) and the tumour uptake of antibodies. METHODS: The affinity and specificity of the cMAb U36 for KAT-4 cells were evaluated in vitro, as was the Na+/I- symporter (NIS) expression. Biodistribution studies were performed on KAT-4 carcinoma-bearing mice injected with 124I-cMAb U36 or free iodine. Biodistribution studies were also performed in animals treated with the specific TGF-beta1 and -beta3 inhibitor Fc:TbetaRII, which lowers TIFP. Treated and non-treated animals were scanned by microPET. RESULTS: Cultured human undifferentiated/anaplastic thyroid carcinoma KAT-4 cells expressed low levels of NIS and uptake of free iodine was insignificant. The cMAb U36 expressed an affinity (KD) of 11+/-2 nM. Tumour radioactivity uptake reached maximum values 48 h after injection of 124I-cMAb U36 (approximately 22%IA/g). KAT-4 carcinomas were readily identified in all 124I-immuno-PET images. Radioactivity tumour uptake in Fc:TbetaRII-treated animals was significantly lower at 24 and 48 h after injection, and five times higher thyroid uptake was also noted. CONCLUSION: We successfully used 124I-cMAb U36 to visualise CD44v6-expressing human anaplastic thyroid carcinoma. Given the lack of NIS expression in KAT-4, tumour visualisation is not due to free iodine uptake. Lowering the TIFP in KAT-4 carcinomas did not increase the uptake of mAbs into tumour tissue.


Subject(s)
Antibodies, Monoclonal/pharmacology , Carcinoma/metabolism , Positron-Emission Tomography/methods , Thyroid Neoplasms/diagnosis , Animals , Antibodies, Monoclonal/chemistry , Cell Differentiation , Cell Line, Tumor , Humans , Hyaluronan Receptors/biosynthesis , Iodine Radioisotopes/pharmacokinetics , Kinetics , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasm Transplantation , Positron-Emission Tomography/instrumentation , Radioimmunoassay/methods , Thyroid Neoplasms/pathology
10.
Int J Cancer ; 119(12): 2795-802, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17019708

ABSTRACT

The stroma of carcinomas shares several characteristics with inflamed tissues including a distorted vasculature, active angiogenesis and macrophage infiltration. In addition, the tumor interstitial fluid pressure (P(IF)) of the stroma is pathologically elevated. We show here that bevacizumab [rhuMab vascular endothelial growth factor (VEGF), Avastin], a monoclonal antibody to VEGF, at a dose of 5 mg/kg modulated inflammation in KAT-4 xenograft human anaplastic thyroid carcinoma tissue. At this dose, bevacizumab reduced the density of macrophages, MHC class II antigen expression by macrophages and IL-1beta mRNA expression. Furthermore, bevacizumab lowered tumor extracellular fluid volume, plasma protein leakage from tumor vessels, the number of CD31-positive structures and tumor P(IF). The tumor plasma volume and the number of alpha-smooth muscle actin-positive vessels, however, remained unchanged. Our data suggest that carcinoma cell-derived VEGF either directly or indirectly participates in maintaining an inflammatory microenvironment in experimental KAT-4 carcinoma. Furthermore, our data indicate that the reduction of inflammation resulting in reduced vascular permeability and decrease in the tumor extracellular fluid volume by bevacizumab contributes to reduced tumor P(IF).


Subject(s)
Antibodies, Monoclonal/therapeutic use , Inflammation/prevention & control , Thyroid Neoplasms/prevention & control , Vascular Endothelial Growth Factor A/immunology , Xenograft Model Antitumor Assays , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Bevacizumab , Blood Vessels/drug effects , Blood Vessels/metabolism , Blood Vessels/pathology , Cell Count , Cell Line, Tumor , Chemokines/genetics , Cytokines/genetics , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Extracellular Fluid/drug effects , Extracellular Fluid/metabolism , Gene Expression/drug effects , Humans , Inflammation/metabolism , Inflammation/pathology , Macrophages/drug effects , Macrophages/pathology , Mice , Mice, SCID , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/prevention & control , Plasma Volume/drug effects , Platelet Endothelial Cell Adhesion Molecule-1/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
11.
Thyroid ; 16(2): 143-50, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16676399

ABSTRACT

Anaplastic thyroid carcinoma (ATC) is one of the most malignant tumors in humans, and currently there is no effective treatment. In the present study we investigated the effect of an endogenous estrogen metabolite, 2-methoxyestradiol (2-ME), on the growth of human ATC cells. 2-ME treatment had a strong growth inhibitory effect on five human ATC cell lines (HTh7, HTh 74, HTh83, C643, and SW1736), but showed no effect on one cell line (KAT-4). Cell cycle analysis of the growth-inhibited cells showed that 2-ME induced a G2/M-arrest, followed by an increased fraction of cells in sub-G1. Analysis of internucleosomal DNA laddering as well as DNA fragmentation in a terminal deoxynucleotide transferase-mediated dUTP nick-end labeling (TUNEL) assay demonstrated a high number of cells undergoing apoptosis after 2-ME treatment. An increased activation of caspase-3 and caspase-8 by 2-ME was observed, and inhibition of caspase-3 decreased the apoptotic effect. Addition of 2-ME increased activity of p38 mitogen-activated protein kinase (MAPK) in the sensitive HTh7 as well as the refractory KAT-4 cells, however, activation of stress-activated protein kinase/c-jun aminoterminal kinase (SAPK/JNK) was seen only in the HTh7 cells. Inhibitors of p38 MAPK and SAPK/JNK significantly attenuated the 2-ME effect. Taken together, our data demonstrate an antiproliferative and apoptotic effect of 2-ME on ATC cells involving activation of MAPKs.


Subject(s)
Apoptosis , Carcinoma/drug therapy , Carcinoma/metabolism , Estradiol/analogs & derivatives , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/metabolism , 2-Methoxyestradiol , Antineoplastic Agents/pharmacology , Blotting, Western , Carcinoma/pathology , Caspase 3 , Caspase 8 , Caspases/metabolism , Cell Line, Tumor , DNA Fragmentation , Estradiol/pharmacology , Flow Cytometry , G1 Phase , Humans , In Situ Nick-End Labeling , MAP Kinase Signaling System , Models, Statistical , Osmosis , RNA, Messenger/metabolism , Ribonucleases/metabolism , Thyroid Neoplasms/pathology , Time Factors , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Biochem Biophys Res Commun ; 338(3): 1625-33, 2005 Dec 23.
Article in English | MEDLINE | ID: mdl-16288728

ABSTRACT

The aim of the present study was to investigate the effect of transforming growth factor-beta1 (TGF-beta1) on telomerase activity in a panel of human anaplastic thyroid carcinoma (ATC) cell lines. Addition of TGF-beta1 decreased the telomerase activity in HTh 74 and KTC-1 cells, while in C 643 and HTh 7 an increased activity was observed. The decreased telomerase activity appeared to be due to transcriptional repression of the hTERT promoter. Addition of a PI-3 kinase inhibitor (LY294002) abrogated the stimulatory effect of TGF-beta1 on the telomerase activity, indicating the possible involvement of hTERT activation via phosphorylation. Furthermore, the MEK-inhibitor U0126 had similar effects suggesting dual regulatory mechanisms. Interestingly, the cell lines differed genetically in that ATC cell lines responding with increased telomerase activity harbored a p53 mutation. In conclusion, TGF-beta1 exerts opposing effects on telomerase activity in ATC cell lines, possibly reflecting deregulation of TGF-beta1 signaling in a more malignant genotype.


Subject(s)
Telomerase/metabolism , Thyroid Neoplasms/enzymology , Transforming Growth Factor beta/pharmacology , Cell Line, Tumor , Down-Regulation/drug effects , Genotype , Humans , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutation/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Promoter Regions, Genetic/genetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-myc/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Telomerase/genetics , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Transforming Growth Factor beta1 , Tumor Suppressor Protein p53/genetics
13.
Lab Invest ; 85(4): 512-21, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15711566

ABSTRACT

A pathologically elevated interstitial fluid pressure (IFP) is a characteristic of both clinical and experimental carcinoma. The soluble TGF-beta receptor type II-murine Fc:IgG2A chimeric protein (Fc:TbetaRII) lowers IFP in the KAT-4 experimental model for anaplastic thyroid carcinoma. Analyses of messenger RNA (mRNA) expressions by Affymetrix microarrays and RNase protection assays, as well as of protein expressions identified tumor macrophages as targets for Fc:TbetaRII. Treatment with Fc:TbetaRII reduced albumin extravasation, increased coverage of alpha-smooth muscle actin-positive cells and reduced expression of NG2, a marker of activated pericytes, in KAT-4 carcinoma blood vessels. Specific inhibition of interleukin-1 (IL-1), a major cytokine produced by activated macrophages, lowered carcinoma IFP to a similar degree as Fc:TbetaRII but had no significant effect on the parameters of blood vessel maturation. Neither Fc:TbetaRII nor inhibition of IL-1 changed blood vessel density. Finally, pretreatment of KAT-4 carcinomas with Fc:TbetaRII increased the antitumor efficacy of doxorubicin. Our data emphasize a potential role of tumor macrophages in carcinoma physiology and identify these cells as potential stromal targets for treatment aimed to improve efficacy of chemotherapy.


Subject(s)
Down-Regulation/physiology , Extracellular Fluid , Macrophages/cytology , Thyroid Neoplasms/pathology , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Doxorubicin/pharmacology , Humans , Immunohistochemistry , Mice , Mice, Nude
14.
Anticancer Res ; 24(6): 3983-90, 2004.
Article in English | MEDLINE | ID: mdl-15736443

ABSTRACT

BACKGROUND: We investigated the effects of 2methoxyestradiol (2-ME), an endogenous estrogenic metabolite, on human endometrial cancer HEC-1-A and RL-95-2 cell lines. MATERIALS AND METHODS: After exposure of HEC-1-A and RL-95-2 cells to 2-ME, the morphological changes were evaluated by acridine orange staining and transmission electron microscopy. Cell cycle progress, apoptosis and necrosis were assessed by flow cytometry, DNA fragmentation and Western blot. RESULTS: 2-ME inhibited cell growth by blocking the S- and G2/M-phase in both cell lines, by inducing apoptosis in HEC-1-A cells and by causing necrosis in RL-95-2 cells. Apoptosis, on HEC-1-A cells, was accompanied by an increased expression of iNOS and STAT1. This apoptotic effect was prevented by the iNOS inhibitor 1400W and eliminated by the caspase inhibitor Z-VAD-FMK. Necrosis, on RL-95-2 cells, was due to a severe disruption of the mitochondrial membrane potential. 2-ME had no significant effect on normal human endometrial cells. CONCLUSION: The data suggest that 2-ME has an antitumor effect on human endometrial carcinoma cells (HEC-1-A and RL-95-2) and may contribute as a new therapeutic agent for endometrial cancer patients.


Subject(s)
Apoptosis/drug effects , Endometrial Neoplasms/drug therapy , Estradiol/analogs & derivatives , Estradiol/pharmacology , 2-Methoxyestradiol , Adult , Cell Cycle/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , DNA-Binding Proteins/biosynthesis , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Endometrial Neoplasms/ultrastructure , Endometrium/cytology , Endometrium/drug effects , Female , Flow Cytometry , Humans , Intracellular Membranes/drug effects , Intracellular Membranes/physiology , Membrane Potentials/drug effects , Middle Aged , Mitochondria/drug effects , Mitochondria/physiology , Necrosis , Nitric Oxide Synthase/biosynthesis , Nitric Oxide Synthase Type II , STAT1 Transcription Factor , Trans-Activators/biosynthesis
15.
Biochem Biophys Res Commun ; 305(4): 1017-23, 2003 Jun 13.
Article in English | MEDLINE | ID: mdl-12767932

ABSTRACT

Mechanism(s) for generation of the high tumor interstitial fluid pressure (TIFP) that is characteristic of carcinoma is not known. We investigated the role of hyaluronan, the major water-binding polysaccharide of the extracellular matrix, for the generation of a high TIFP. A human anaplastic thyroid carcinoma (KAT-4) xenografted to athymic mice and a syngeneic rat colon carcinoma (PROb) were used. Neither KAT-4 nor PROb cells produced hyaluronan (HA) in culture, however, both cell lines produced factors that stimulated HA-synthesis by cultured fibroblasts. Modulating hyaluronan levels by transfection of PROb carcinoma cells with hyaluronan synthase-2 revealed no correlation between hyaluronan content and TIFP. Furthermore, lowering of TIFP by treating KAT-4 tumors with a specific inhibitor of TGF-beta 1 and -beta 3 did not change the concentration of hyaluronan in the tumors. In summary, our results suggest that a modulation of hyaluronan content is not a major pathogenetic mechanism for the generation of the characteristically high TIFP in malignant carcinomas.


Subject(s)
Carcinoma/chemistry , Hyaluronic Acid/analysis , Animals , Carcinoma/metabolism , Carcinoma/pathology , Cell Line , Colorectal Neoplasms/chemistry , Colorectal Neoplasms/metabolism , Extracellular Space , Female , Fibroblasts/metabolism , Humans , Hyaluronic Acid/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Nude , Pressure , Rats , Thyroid Neoplasms/chemistry , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta1 , Transforming Growth Factor beta3 , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
16.
Mol Biol Cell ; 14(2): 529-44, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12589052

ABSTRACT

The inhibitory Smad7, a direct target gene for transforming growth factor-beta (TGF-beta), mediates TGF-beta1-induced apoptosis in several cell types. Herein, we report that apoptosis of human prostate cancer PC-3U cells induced by TGF-beta1 or Smad7 overexpression is caused by a specific activation of the p38 mitogen-activated protein kinase pathway in a TGF-beta-activated kinase 1 (TAK1)- and mitogen-activated protein kinase kinase 3 (MKK3)-dependent manner. Expression of dominant negative p38, dominant negative MKK3, or incubation with the p38 selective inhibitor [4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-5-(4-pyridyl)1H-imidazole], prevented TGF-beta1-induced apoptosis. The expression of Smad7 was required for TGF-beta-induced activation of MKK3 and p38 kinases, and endogenous Smad7 was found to interact with phosphorylated p38 in a ligand-dependent manner. Ectopic expression of wild-type TAK1 promoted TGF-beta1-induced phosphorylation of p38 and apoptosis, whereas dominant negative TAK1 reduced TGF-beta1-induced phosphorylation of p38 and apoptosis. Endogenous Smad7 was found to interact with TAK1, and TAK1, MKK3, and p38 were coimmunoprecipitated with Smad7 in transiently transfected COS1 cells. Moreover, ectopically expressed Smad7 enhanced the coimmunoprecipitation of HA-MKK3 and Flag-p38, supporting the notion that Smad7 may act as a scaffolding protein and facilitate TAK1- and MKK3-mediated activation of p38.


Subject(s)
Apoptosis , DNA-Binding Proteins/metabolism , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Prostatic Neoplasms/pathology , Protein-Tyrosine Kinases/metabolism , Trans-Activators/metabolism , Transforming Growth Factor beta/physiology , Animals , Blotting, Western , COS Cells , DNA Fragmentation , Enzyme Activation , Enzyme Inhibitors/pharmacology , Genes, Dominant , Humans , In Situ Nick-End Labeling , MAP Kinase Kinase 3 , Male , Microscopy, Fluorescence , Phosphorylation , Precipitin Tests , Protein Binding , Smad7 Protein , Time Factors , Transfection , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1 , Tumor Cells, Cultured , p38 Mitogen-Activated Protein Kinases
17.
Thyroid ; 12(9): 781-90, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12481943

ABSTRACT

Loss of the epithelial phenotype is a well-established phenomenon during progression of carcinomas to a more malignant state. In the present study, we describe a human thyroid tumor cell line (KAT-4), established from a poorly differentiated carcinoma, which displays exceptional features. In culture, the KAT-4 cells had a fast proliferation rate that was not restricted by high cell density, resulting in multilayered growth. Unexpectedly, the cells expressed normal levels of epithelial markers, e.g., cytokeratin, occludin, and E-cadherin, showed apical-basolateral polarization of the plasma membrane including microvilli and junction complexes, and formed intercellular lumens resembling thyroid follicles. Yet, when grown on filter, the cells were unable to establish a tight paracellular barrier. Moreover, E-cadherin expressed at the cell surface consisted of two peptides with abnormal size (135 and 95 kd, respectively) as compared to mature E-cadherin (120 kd) in nonneoplastic thyrocytes. Northern blot analysis and examination of immunoreactivity, glycosylation, and catenin binding suggested that E-cadherin was aberrant because of altered posttranscriptional processing. Thus, the KAT-4 thyroid carcinoma cell line has a unique phenotype, with maintained epithelial morphology despite dysfunctioning tight junctions, abnormal E-cadherin, and loss of contact-inhibited growth, that is not previously identified in other wild-type tumor cell lines.


Subject(s)
Cadherins/metabolism , Carcinoma, Giant Cell , Epithelial Cells/metabolism , Epithelial Cells/pathology , Thyroid Neoplasms , Aged , Aged, 80 and over , Biological Transport , Cadherins/genetics , Cell Division , Contact Inhibition , Epithelial Cells/ultrastructure , Female , Humans , Microscopy, Electron , RNA Processing, Post-Transcriptional , Tight Junctions/metabolism , Tight Junctions/pathology , Tight Junctions/ultrastructure , Tumor Cells, Cultured
18.
FEBS Lett ; 531(2): 141-51, 2002 Nov 06.
Article in English | MEDLINE | ID: mdl-12417302

ABSTRACT

Prostate and breast carcinomas are sex hormone-related carcinomas, which are known to be associated with an over-expression of the proto-oncogene Bcl-2. Here, we report that 2-methoxyestradiol (2-ME), an endogenous metabolite of estrogen that does not bind to nuclear estrogen receptors, effectively induces apoptosis in Bcl-2-expressing human prostate and breast carcinoma cells in vitro and in a rat prostate tumor model in vivo. In several cell lines derived from prostate, breast, liver and colorectal carcinomas, 2-ME treatment led to an activation of c-Jun N-terminal kinase (JNK) and phosphorylation of Bcl-2, which preceded the induction of apoptosis. In summary, our data suggest that 2-ME induces apoptosis in epithelial carcinomas by causing phosphorylation of JNK, which appears to be correlated with phosphorylation of Bcl-2.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Carcinoma/metabolism , Estradiol/analogs & derivatives , Estradiol/pharmacology , Prostatic Neoplasms/metabolism , 2-Methoxyestradiol , Animals , Breast/cytology , Breast/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma/pathology , Caspases/physiology , Cell Division/drug effects , Colonic Neoplasms/pathology , Endothelium/cytology , Endothelium/drug effects , Female , Humans , Kinetics , Liver Neoplasms/pathology , Male , Mitogen-Activated Protein Kinases/metabolism , Pertussis Toxin/pharmacology , Phosphorylation/drug effects , Prostatic Neoplasms/pathology , Proto-Oncogene Mas , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Inbred F344 , Tumor Cells, Cultured
19.
Int J Cancer ; 102(5): 453-62, 2002 Dec 10.
Article in English | MEDLINE | ID: mdl-12432546

ABSTRACT

A high tumor interstitial fluid pressure (TIFP) is a pathologic characteristic distinguishing the stroma of carcinomas from normal interstitial loose connective tissues. The role of TGF-beta1 and -beta3 in generating a high TIFP was investigated in xenografted experimental anaplastic thyroid carcinoma (ATC) derived from the human ATC cell line KAT-4. A single intravenous injection of a soluble recombinant TGF-beta receptor type II-murine Fc:IgG(2A) chimeric protein that specifically inhibits TGF-beta1 and -beta3, significantly lowered TIFP in a time and concentration dependent manner but did not change total tissue water content in the tumors. Tumor growth rate was higher in tumors treated with the TGF-beta1 and -beta3 inhibitor compared to control tumors during the first 10 days after administration of the inhibitor. The apoptotic index of carcinoma cells, and expression of the cell cycle inhibitor p27(Kip1), were, however, increased in TGF-beta1 and -beta3 inhibitor-treated tumors. Prolonged treatment periods and administration of a second dose of the inhibitor decreased tumor growth rate. The TGF-beta1 and -beta3 inhibitor did not affect proliferation or expression of phosphorylated Smad2 protein in KAT-4 cells cultured in vitro. Our results indicate that members of the TGF-beta family are potential targets for novel anti-cancer treatment directed to the stroma. First by controlling TIFP and by that potentially the uptake of anticancer drugs into tumors and second by their suggested role in maintaining a supportive tumor stroma.


Subject(s)
Carcinoma/metabolism , Extracellular Space/physiology , Thyroid Neoplasms/metabolism , Transforming Growth Factor beta/metabolism , Animals , Apoptosis , Carcinoma/pathology , Cell Cycle Proteins/metabolism , Cell Division , Cyclin-Dependent Kinase Inhibitor p27 , Humans , Mice , Neoplasm Transplantation , Pressure , Protein Serine-Threonine Kinases , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta , Recombinant Fusion Proteins , Stromal Cells/metabolism , Thyroid Neoplasms/pathology , Transforming Growth Factor beta1 , Transforming Growth Factor beta3 , Transplantation, Heterologous , Tumor Cells, Cultured , Tumor Suppressor Proteins/metabolism
20.
J Cell Sci ; 115(Pt 22): 4227-36, 2002 Nov 15.
Article in English | MEDLINE | ID: mdl-12376555

ABSTRACT

Enhancement of tumor cell growth and invasiveness by transforming growth factor-beta (TGF-beta) requires constitutive activation of the ras/MAPK pathway. Here we have investigated how MEK activation by epidermal growth factor (EGF) influences the response of fully differentiated and growth-arrested pig thyroid epithelial cells in primary culture to TGF-beta1. The epithelial tightness was maintained after single stimulation with EGF or TGF-beta1 (both 10 ng/ml) for 48 hours. In contrast, co-stimulation abolished the transepithelial resistance and increased the paracellular flux of [(3)H]inulin within 24 hours. Reduced levels of the tight junction proteins claudin-1 and occludin accompanied the loss of barrier function. N-cadherin, expressed only in few cells of untreated or single-stimulated cultures, was at the same time increased 30-fold and co-localised with E-cadherin at adherens junctions in all cells. After 48 hours of co-stimulation, both E- and N-cadherin were downregulated and the cells attained a fibroblast-like morphology and formed multilayers. TGF-beta1 only partially inhibited EGF-induced Erk phosphorylation. The MEK inhibitor U0126 prevented residual Erk phosphorylation and abrogated the synergistic responses to TGF-beta1 and EGF. The observations indicate that concomitant growth factor-induced MEK activation is necessary for TGF-beta1 to convert normal thyroid epithelial cells to a mesenchymal phenotype.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Epidermal Growth Factor/metabolism , Epithelial Cells/enzymology , Mesoderm/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Neoplasm Invasiveness/genetics , Protein Serine-Threonine Kinases/metabolism , Transforming Growth Factor beta/metabolism , Adherens Junctions/drug effects , Adherens Junctions/metabolism , Adherens Junctions/ultrastructure , Animals , Cadherins/drug effects , Cadherins/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Size/drug effects , Cell Size/physiology , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Claudin-1 , Down-Regulation/drug effects , Down-Regulation/physiology , Drug Interactions/physiology , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/ultrastructure , MAP Kinase Kinase 1 , Membrane Proteins/drug effects , Membrane Proteins/metabolism , Mesoderm/drug effects , Mesoderm/ultrastructure , Microscopy, Electron , Mitogen-Activated Protein Kinase Kinases/drug effects , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Invasiveness/physiopathology , Occludin , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/genetics , Sus scrofa , Thyroid Gland/cytology , Thyroid Gland/drug effects , Thyroid Gland/enzymology , Tight Junctions/drug effects , Tight Junctions/metabolism , Tight Junctions/ultrastructure , Transforming Growth Factor beta/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...