Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Cell Mol Med ; 11(4): 826-38, 2007.
Article in English | MEDLINE | ID: mdl-17760843

ABSTRACT

Adhesion molecules of the integrin beta1 family are thought to be involved in the malignant progression renal cell carcinoma (RCC). Still, it is not clear how they contribute to this process. Since the hematogenous phase of tumour dissemination is the rate-limiting step in the metastatic process, we explored beta1 integrin alterations on several RCC cell lines (A498, Caki1, KTC26) before and after contacting vascular endothelium in a tumour-endothelium (HUVEC) co-culture assay. Notably, alpha2, alpha3 and alpha5 integrins became down-regulated immediately after the tumour cells attached to HUVEC, followed by re-expression shortly thereafter. Integrin down-regulation on RCC cells was caused by direct contact with endothelial cells, since the isolated endothelial membrane fragments but not the cell culture supernatant contributed to the observed effects. Integrin loss was accompanied by a reduced focal adhesion kinase (FAK) expression, FAK activity and diminished binding of tumour cells to matrix proteins. Furthermore, intracellular signalling proteins RCC cells were altered in the presence of HUVEC membrane fragments, in particular 14-3-3 epsilon, ERK2, PKCdelta, PKCepsilon and RACK1, which are involved in regulating tumour cell motility. We, therefore, speculate that contact of RCC cells with the vascular endothelium converts integrin-dependent adhesion to integrin-independent cell movement. The process of dynamic integrin regulation may be an important part in tumour cell migration strategy, switching the cells from being adhesive to becoming motile and invasive.


Subject(s)
Cell Membrane/metabolism , Down-Regulation , Endothelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Integrin beta1/metabolism , Kidney Neoplasms/metabolism , Cell Adhesion , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Endothelial Cells/enzymology , Flow Cytometry , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Integrin beta1/genetics , Kidney Neoplasms/genetics , Microscopy, Confocal , Signal Transduction , Subcellular Fractions/metabolism , Umbilical Cord/cytology , Umbilical Cord/enzymology
2.
Exp Cell Res ; 313(19): 4051-65, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17706641

ABSTRACT

The mechanisms leading to renal cell carcinoma (RCC) metastasis are incompletely understood. Although evidence shows that the chemokine receptor CXCR4 and its ligand CXCL12 may regulate tumor dissemination, their role in RCC is not clearly defined. We examined CXCR4 expression and functionality on RCC cell lines, and explored CXCL12-triggered tumor adhesion to human endothelium (HUVEC) or extracellular matrix proteins. Functional CXCR4 was expressed on A498 tumor cells, enabling them to migrate towards a CXCL12 gradient. CXCR4 engagement by CXCL12 induced elevated cell adhesion to HUVEC, to immobilized fibronectin, laminin or collagen. Anti-CXCR4 antibodies or CXCR4 knock down by siRNA applied prior to CXCL12 stimulation impaired CXCL12-triggered tumor adhesion. However, blocking CXCR4 subsequent to CXCL12 stimulation did not. This pointed to an indirect control of tumor cell adhesion by CXCR4. In fact, CXCR4 engagement by CXCL12 also induced alterations of receptors of the integrin family, notably alpha3, alpha5, beta1 and beta3 subunits, and blocking beta1 integrins with a function-blocking antibody prevented CXCL12-induced A498 adhesion. Focal adhesion kinase (total and activated) and integrin-linked kinase significantly increased in CXCL12-treated A498 cells, accompanied by a distinct up-regulation of ERK1/2, JNK and p38 phosphorylation. Therefore, CXCR4 may be crucial in controlling adhesion of A498 cells via cross talking with integrin receptors. These data show that CXCR4 receptors contribute to RCC dissemination and may provide a novel link between CXCR4 chemokine receptor expression and integrin triggered RCC adhesion to the vascular wall and subendothelial matrix components.


Subject(s)
Carcinoma, Renal Cell/pathology , Cell Adhesion , Chemokine CXCL12/physiology , Integrins/metabolism , Receptors, CXCR4/metabolism , Cell Line, Tumor , Chemotaxis , Coculture Techniques , Endothelium, Vascular/cytology , Humans , Integrin beta1/metabolism , Neoplasm Metastasis
3.
BMC Cancer ; 6: 294, 2006 Dec 21.
Article in English | MEDLINE | ID: mdl-17181871

ABSTRACT

BACKGROUND: Drug resistance to chemotherapy is often associated with increased malignancy in neuroblastoma (NB). One explanation for the link between resistance and malignancy might be that resistance facilitates cancer progression and invasion. To investigate this hypothesis, adhesion, transendothelial penetration and NCAM (CD56) adhesion receptor expression of drug-resistant versus drug-sensitive NB tumor cells were evaluated. METHODS: Acquired drug resistance was mimicked by exposing parental UKF-NB-2, UKF-NB-3 or IMR-32 tumor cells to increasing concentrations of vincristine- (VCR) or doxorubicin (DOX) to establish the resistant tumor cell sublines UKF-NB-2VCR, UKF-NB-2DOX, UKF-NB-3VCR, UKF-NB-3DOX, IMR-32VCR and IMR-32DOX. Additionally, the malignant behaviour of UKF-NB-4, which already possessed the intrinsic multidrug resistance (MDR) phenotype, was analyzed. UKF-NB-4 exposed to VCR or DOX were designated UKF-NB-4VCR or UKF-NB-4DOX. Combined phase contrast - reflection interference contrast microscopy was used to separately evaluate NB cell adhesion and penetration. NCAM was analyzed by flow cytometry, western blot and RT-PCR. RESULTS: VCR and DOX resistant tumor sublines showed enhanced adhesion and penetration capacity, compared to their drug naïve controls. Strongest effects were seen with UKF-NB-2VCR, UKF-NB-3VCR and IMR-32DOX. DOX or VCR treatment also evoked increased invasive behaviour of UKF-NB-4. The process of accelerated tumor invasion was accompanied by decreased NCAM surface and protein expression, and down-regulation of NCAM coding mRNA. Transfection of UKF-NB-4VCR cells with NCAM cDNA led to a significant receptor up-regulation, paralleled by diminished adhesion to an endothelial cell monolayer. CONCLUSION: It is concluded that NB cells resistant to anticancer drugs acquire increased invasive capacity relative to non-resistant parental cells, and that enhanced invasion is caused by strong down-regulation of NCAM adhesion receptors.


Subject(s)
CD56 Antigen/metabolism , Cell Adhesion , Cell Movement , Drug Resistance, Neoplasm , Endothelial Cells/metabolism , Neural Cell Adhesion Molecules/metabolism , Neuroblastoma/immunology , Antineoplastic Agents/pharmacology , Blotting, Western , CD56 Antigen/genetics , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation , Down-Regulation , Doxorubicin/pharmacology , Drug Resistance, Multiple , Flow Cytometry , Humans , Microscopy, Interference , Neoplasm Invasiveness , Neural Cell Adhesion Molecules/genetics , Neuroblastoma/pathology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transfection , Vincristine/pharmacology
4.
Neoplasia ; 8(10): 807-16, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17032497

ABSTRACT

The genome and antigens of human cytomegalovirus (HCMV) are frequently found in prostatic carcinoma. However, whether this infection is causative or is an epiphenomenon is not clear. We therefore investigated the ability of HCMV to promote metastatic processes, defined by tumor cell adhesion to the endothelium and extracellular matrix proteins. Experiments were based on the human prostate tumor cell line PC3, either infected with the HCMV strain Hi (HCMV(Hi)) or transfected with cDNA encoding the HCMV-specific immediate early protein IEA1 (UL123) or IEA2 (UL122). HCMV(Hi) upregulated PC3 adhesion to the endothelium and to the extracellular matrix proteins collagen, laminin, and fibronectin. The process was accompanied by enhancement of beta(1)-integrin surface expression, elevated levels of integrin-linked kinase, and phosphorylation of focal adhesion kinase. IEA1 or IEA2 did not modulate PC3 adhesion or beta(1)-integrin expression. Based on this in vitro model, we postulate a direct association between HCMV infection and prostate tumor transmigration, which is not dependent on IEA proteins. Integrin overexpression, combined with the modulation of integrin-dependent signalling, seems to be, at least in part, responsible for a more invasive PC3(Hi) tumor cell phenotype. Elevated levels of c-myc found in IEA1-transfected or IEA2-transfected PC3 cell populations might promote further carcinogenic processes through accelerated cell proliferation.


Subject(s)
Carcinoma/pathology , Carcinoma/virology , Cell Adhesion , Cytomegalovirus Infections , Prostatic Neoplasms/pathology , Prostatic Neoplasms/virology , Antigens , Antigens, Neoplasm , Gene Expression Regulation, Neoplastic , Humans , Integrin beta1/metabolism , Male , Neoplasm Invasiveness , Neoplasm Metastasis , Phenotype , Proto-Oncogene Proteins c-myc/metabolism , Tumor Cells, Cultured
5.
Int J Mol Med ; 18(2): 347-54, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16820945

ABSTRACT

Renal cell carcinoma (RCC) is the most common malignant tumor of the kidney. Adhesion receptors of the beta1 integrin family are assumed to be involved in carcinogenesis, but it is not clear how they contribute to RCC progression. In an in vitro model, we evaluated growth and adhesion capacity of Caki-I and KTC-26 kidney carcinoma cell lines compared to normal renal proximal tubular epithelial cells (PTC). alpha1-alpha6beta1 integrin subunits in malignant and non-malignant cells were evaluated by Western blotting and RT-PCR, integrin surface expression was measured by flow cytometry and confocal microscopy. Additionally, tumor cells were allowed to re-differentiate in the presence of valproic acid (VPA) and dynamic alterations of the integrin profile were analyzed. Caki-I and KTC-26 were characterized by accelerated proliferation and adhesion to an endothelial cell monolayer, compared to PTC cells. The integrin beta1 repertoire in RCC cell lines was significantly different from that detected in PTC, and included down-regulated alpha2 and alpha6, but up-regulated alpha1, alpha3 and alpha5 proteins. VPA application reduced tumor malignancy which was evidenced by reduced cell growth and adhesion capacity. The reduction in tumor malignancy was paralleled by the integrin expression profile of renal tumor cells 'matching' the pattern seen in PTC. We assume that a sensitive integrin balance exists in normal renal epithelial cells, and that dysregulation of the 'physiological' receptor equipment drives these cells towards malignancy. VPA acted on all investigated integrin subtypes and restored the receptor pattern typical for non-malignant cells. Therefore, VPA may represent a novel therapeutic option in RCC treatment.


Subject(s)
Carcinoma, Renal Cell/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor/drug effects , Enzyme Inhibitors/pharmacology , Integrin beta1/metabolism , Kidney Neoplasms/metabolism , Valproic Acid/pharmacology , Cell Adhesion/physiology , Cell Line, Tumor/physiology , Cells, Cultured , Disease Progression , Dose-Response Relationship, Drug , Epithelial Cells/cytology , Epithelial Cells/metabolism , Humans , Integrin alpha Chains/genetics , Integrin alpha Chains/metabolism , Integrin beta1/genetics , Kidney Tubules, Proximal/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...