Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
Oncogene ; 37(7): 897-911, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29059159

ABSTRACT

Malignant melanoma reveals rapidly increasing incidence and mortality rates worldwide. By now, BRAF inhibition is the standard therapy for advanced melanoma in patients carrying BRAF mutations. However, only approximately 50% of melanoma patients harbor therapeutically attackable BRAF mutations, and overall survival after treatment with BRAF inhibitors is modest. KRAS (Kirsten Rat sarcoma) proteins are acting upstream of BRAF and have a major role in human cancer. Recent approaches awaken the hope to use KRAS inhibition (KRASi) as a clinical tool. In this study, we identified wild-type KRAS as a novel therapeutic target in melanoma. KRASi functions synergistically with BRAF inhibition to reduce melanoma proliferation and to induce apoptosis independently of BRAF mutational status. Moreover, acquired resistance to BRAF inhibitors in melanoma is dependent on dynamic regulation of KRAS expression with subsequent AKT and extracellular-signal regulated kinase activation and can be overcome by KRASi. This suggests KRASi as novel approach in melanoma-alone or in combination with other therapeutic regimes.


Subject(s)
Biomarkers, Tumor/metabolism , Drug Resistance, Neoplasm/genetics , Melanoma/pathology , Mutation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/genetics , Skin Neoplasms/pathology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Follow-Up Studies , Humans , Male , Melanoma/drug therapy , Melanoma/genetics , Mice , Mice, Nude , Prognosis , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Signal Transduction , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Melanoma, Cutaneous Malignant
2.
Environ Mol Mutagen ; 58(3): 172-177, 2017 04.
Article in English | MEDLINE | ID: mdl-28370324

ABSTRACT

Cold atmospheric argon plasma is recognized as a new contact free approach for the decrease of bacterial load on chronic wounds in patients. So far very limited data are available on its toxicity and mutagenicity on eukaryotic cells. Thus, the toxic/mutagenic potential of cold atmospheric argon plasma using the MicroPlaSter ß® , which has been used efficiently in humans treating chronic and acute wounds, was investigated using the XTT assay in keratinocytes and fibroblasts and the HGPRT (hypoxanthine guanine phosphoribosyl transferase) assay with V79 Chinese hamster cells. The tested clinical parameter of a 2 min cold atmospheric argon plasma treatment revealed no relevant toxicity on keratinocytes (viability: 76% ± 0.17%) and on fibroblasts (viability: 81.8 ± 0.10) after 72 hr as compared to the untreated controls. No mutagenicity was detected in the HGPRT assay with V79 cells even after repetitive CAP treatments of 2-10 min every 24 hr for up to 5 days. In contrast, UV-C irradiation of V79 cells, used as a positive control in the HGPRT test, led to DNA damage and mutagenic effects. Our findings indicate that cold atmospheric plasma using the MicroPlaSter ß® shows negligible effects on keratinocytes and fibroblasts but no mutagenic potential in the HGPRT assay, indicating a new contact free safe technology. Environ. Mol. Mutagen. 58:172-177, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Argon/toxicity , Fibroblasts/drug effects , Keratinocytes/drug effects , Mutagens/toxicity , Plasma Gases/toxicity , Animals , Cell Survival/drug effects , Cricetinae , Fibroblasts/pathology , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Keratinocytes/pathology , Mutagenicity Tests , Primary Cell Culture
3.
Br J Cancer ; 109(12): 3116-24, 2013 Dec 10.
Article in English | MEDLINE | ID: mdl-24169347

ABSTRACT

BACKGROUND: Processing of microRNAs (miRNAs) is a highly controlled process. Deregulation of miRNA expression was observed in several types of cancer but changes in the miRNA-processing enzymes have not been analysed until today. In this study, we analysed Argonaute2 (AGO2, EIF2C2), as one main factor of the miRNA processing ensemble, in the context of cancer development, especially in melanoma. METHODS: We determined the AGO2 expression level in melanoma, as well as in other cancers, with biochemical approaches (qRT-PCR, western blot and immunofluorescence studies) and analysed the cell behaviour in migration assays. RESULTS: Specifically in melanoma, we revealed a strong reduction of AGO2 expression compared with primary melanocytes. The reduction of AGO2 expression was only found on protein level, whereas the mRNA level stayed unchanged hinting to post-transcriptional regulation. We could show that re-expression of AGO2 in melanoma leads to a strong improvement of regulatory effects due to increased functionality of small-interfering RNAs and short hairpin RNAs. CONCLUSION: We identified melanoma-specific downregulation of AGO2 and corresponding reduced RNAi efficiency. These findings will help to understand the molecular basis of malignant melanoma and can potentially lead to an improvement of therapeutic strategies.


Subject(s)
Argonaute Proteins/deficiency , Argonaute Proteins/genetics , Melanoma/metabolism , MicroRNAs/genetics , Argonaute Proteins/biosynthesis , Argonaute Proteins/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , Melanoma/genetics , Melanoma/pathology , MicroRNAs/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Transfection
4.
Br J Cancer ; 108(7): 1460-9, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23511560

ABSTRACT

BACKGROUND: The role of melanoma inhibitory activity 2 (MIA2) was examined in human oral squamous cell carcinoma (OSCC). METHODS: MIA2 role was examined by immunohistochemistry of human OSCCs and knockdown studies using human 3 OSCC cell lines with MIA2 expression. RESULTS: MIA2 expression was observed in 62 (66.7%) of 93 OSCCs and was associated with tumour expansion and nodal metastasis. Melanoma inhibitory activity 2 expression was inversely correlated with intratumoral infiltration of lymphocytes. Invasion and anti-apoptotic survival were reduced by MIA2 knockdown in HSC3 cells. MOLT-3 lymphocytes infiltrating the HSC3 cell layer was enhanced by MIA2 knockdown or MIA2 depletion with the antibody. In HSC3 cells, MIA2 knockdown decreased the expressions of vascular endothelial growth factor (VEGF), VEGF-C, and VEGF-D. The downregulation of VEGF-C and -D was caused by inhibition of p38 and extracellular signal-regulated kinase (ERK)1/2, respectively. Melanoma inhibitory activity 2 was co-precipitated with integrin α4 andα5 in HSC3 cells. Integrin α4 knockdown decreased p38 phosphorylation and increased apoptosis, whereas integrin α5 knockdown decreased c-Jun N-terminal kinase (JNK) phosphorylation and apoptosis. Inhibition of JNK decreased apoptosis in the HSC3 cells. CONCLUSION: These findings suggest that the roles of MIA2 might be based on the variety of the integrins and the subtypes of mitogen-activated protein kinase.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Mouth Neoplasms/metabolism , Tumor Suppressor Proteins/metabolism , Aged , Antigens, Neoplasm , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Female , Gene Knockdown Techniques , Humans , Immunohistochemistry , Integrins/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Male , Middle Aged , Mitogen-Activated Protein Kinases/metabolism , Mouth Neoplasms/immunology , Mouth Neoplasms/pathology , Neoplasm Proteins , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/immunology , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor C/biosynthesis , Vascular Endothelial Growth Factor D/biosynthesis
5.
Oncogene ; 32(7): 837-48, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-22469980

ABSTRACT

Bone morphogenetic proteins (BMPs) are known to play an important role in melanoma development and progression. However, the downstream targets of BMPs have not been investigated thus far. Therefore, we treated melanoma cell lines with the Smad-specific BMP inhibitor Dorsomorphin and performed a cDNA microarray. We identified death inducer-obliterator 1 (Dido1) as a BMP-specific Smad-regulated target gene, which was confirmed by qRT-PCR, immunofluorescence staining and electrophoretic mobility shift assay experiments. An analysis of Dido1 expression revealed an upregulation of Dido1 levels in melanoma cell lines and tissues compared with normal melanocytes. Colony-formation assays showed that siDido1-transfected cells formed significantly smaller colonies when grown in soft agar compared with control cells. In addition, fluorescence-activated cell sorting and western blot experiments revealed that transfection of melanoma cells with Dido1 small interfering RNAs led to an upregulation of apoptosis. Furthermore, cell migratory and invasive potentials were strongly reduced in siDido1-transfected cells compared with control cells. Finally, we demonstrated that Dido1 induces the expression of Integrin αV, thereby promoting the attachment, migration, invasion and apoptosis resistance of melanoma cells.


Subject(s)
Bone Morphogenetic Proteins/pharmacology , DNA-Binding Proteins/physiology , Gene Expression Regulation, Neoplastic/drug effects , Melanoma/genetics , Skin Neoplasms/genetics , Apoptosis/drug effects , Apoptosis/genetics , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/physiology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Progression , Gene Expression Profiling , Humans , Integrin alphaV/genetics , Integrin alphaV/metabolism , Melanoma/pathology , Microarray Analysis , Neoplasm Invasiveness , RNA, Small Interfering/pharmacology , Skin Neoplasms/pathology , Smad4 Protein/antagonists & inhibitors , Smad4 Protein/genetics
6.
Oncogene ; 32(24): 2984-91, 2013 Jun 13.
Article in English | MEDLINE | ID: mdl-22797068

ABSTRACT

A fundamental event in the development and progression of malignant melanoma is the deregulation of cancer-relevant transcription factors. We recently showed that c-Jun is a main regulator of tumor progression in melanoma and thus the most important member of the AP-1 transcription factor family for this disease. Interestingly, we revealed that c-Jun expression was regulated on the post-transcriptional level and therefore speculated that miRNAs could be involved in c-Jun regulation. We determined seed sequences for miR-125b and miR-527 in the coding region of c-Jun mRNA that hints at the direct involvement of miRNA-dependent regulation on the protein level. We found that the expression of miR-125b was significantly reduced in malignant melanoma cell lines and tissue samples compared with melanocytes, whereas miR-527 remained unchanged. In further functional experiments, treatment of melanoma cells with pre-miR-125b resulted in strong suppression of cellular proliferation and migration, supporting the role of miR-125b in melanoma. In addition, transfection of pre-miR-125b led to strong downregulation of c-Jun protein but not mRNA expression in melanoma cells. Luciferase assays using reporter plasmids containing the miR-125b seed sequence in the luciferase coding region confirmed the direct interaction with miR-125b. Furthermore, immunoprecipitation of Ago-2 revealed that c-Jun mRNA accumulated in the RNA-induced silencing complex after pre-miR-125b transfection in melanoma cells. In summary, we identified an important role for miR-125b in malignant melanoma. Moreover, we demonstrated post-transcriptional regulation of c-Jun by this miRNA and showed that c-Jun is a main mediator of the effects of miR-125b on melanoma cells.


Subject(s)
Disease Progression , Gene Expression Regulation, Neoplastic/genetics , Melanoma/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Base Sequence , Cell Line, Tumor , Humans , Melanoma/metabolism , Melanoma/pathology , Transcription, Genetic/genetics
7.
Br J Cancer ; 105(2): 231-8, 2011 Jul 12.
Article in English | MEDLINE | ID: mdl-21673687

ABSTRACT

BACKGROUND: Malignant melanoma cells are known to have altered expression of growth factors compared with normal human melanocytes. These changes most likely favour tumour growth and progression, and influence tumour environment. The induction of transforming growth factor beta1, 2 and 3 as well as BMP4 and BMP7 expression in malignant melanoma has been reported before, whereas the expression of an important modulator of these molecules, connective tissue growth factor (CTGF), has not been investigated in melanomas until now. METHODS: Expression of CTGF was analysed in melanoma cell lines and tissue samples by qRT-PCR and immunohistochemistry. To determine the regulation of CTGF expression in malignant melanoma, specific siRNA was used. Additionally, migration, invasion and attachment assays were carried out. RESULTS: We were able to demonstrate that CTGF expression is upregulated in nine melanoma cell lines and in primary and metastatic melanoma in situ. The transcription factor HIF-1α was revealed as a positive regulator for CTGF expression. Melanoma cells, in which CTGF expression is diminished, show a strong reduction of migratory and invasive properties when compared with controls. Further, treatment of normal human epidermal melanocytes with recombinant CTGF leads to an increase of migratory and invasive behaviour of these cells. CONCLUSION: These results suggest that CTGF promotes melanoma cell invasion and migration and, therefore, has an important role in the progression of malignant melanoma.


Subject(s)
Cell Movement/genetics , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/physiology , Melanoma/genetics , Melanoma/pathology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Protein 7/metabolism , Cell Line, Tumor , Cells, Cultured , Connective Tissue Growth Factor/antagonists & inhibitors , Connective Tissue Growth Factor/pharmacology , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Melanocytes/drug effects , Melanocytes/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neoplasm Invasiveness , RNA, Small Interfering/pharmacology , Up-Regulation/drug effects , Up-Regulation/genetics
8.
Oncogene ; 30(44): 4523-30, 2011 Nov 03.
Article in English | MEDLINE | ID: mdl-21577203

ABSTRACT

CYLD is a deubiquitination enzyme that regulates different cellular processes, such as cell proliferation and cell survival. Mutation and loss of heterozygosity of the CYLD gene causes development of cylindromatosis, a benign tumour originating from the skin. Our study shows that CYLD expression is dramatically downregulated in basal cell carcinoma (BCC), the most common cancer in humans. Reduced CYLD expression in basal cell carcinoma was mediated by GLI1-dependent activation of the transcriptional repressor Snail. Inhibition of GLI1 restored the CYLD expression-mediated Snail signaling pathway, and caused a significant delay in the G1 to S phase transition, as well as proliferation. Our data suggest that GLI1-mediated suppression of CYLD has a significant role in basal cell carcinoma progression.


Subject(s)
Gene Expression Regulation, Neoplastic , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Carcinoma, Basal Cell/genetics , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Deubiquitinating Enzyme CYLD , Humans , Skin Neoplasms/genetics , Snail Family Transcription Factors , Zinc Finger Protein GLI1
9.
Carcinogenesis ; 32(6): 935-43, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21459757

ABSTRACT

Slits are a group of secreted glycoproteins that act as molecular guidance cues in cellular migration. Recently, several studies demonstrated that Slit-2 can operate as candidate tumour suppressor protein in various tissues. In this study, we show Slit-2 expression in basal cell layers of normal oral mucosa colocalized with P-cadherin expression. In contrast, there is a loss of Slit-2 and P-cadherin expression in mucosa of oral squamous cell carcinoma (OSCC). Our in vitro investigations reveal a correlation of P-cadherin and Slit-2 expression: OSCC cells with induced P-cadherin expression (PCI52_PC) display an increased Slit-2 expression. However, abrogating P-cadherin function with a function-blocking antibody decreases Slit-2 secretion confirming a direct link between P-cadherin and Slit-2. Moreover, experiments with OSCC cells show that Slit-2 interferes with a Wnt related signalling pathway, which in turn affects Slit-2 expression in a feedback loop. Functionally, transwell migration assays demonstrate a Slit-2 dose-dependent decrease of PCI52_PC cell migration. However, there is no influence on migration in mock control cells. Responsible for this migration block might be an interaction of P-cadherin with Roundabout (Robo)-3, a high affinity receptor of Slit-2. Indeed, proximity ligation assays exhibit P-cadherin/Robo-3 interactions on PCI52_PC cells. Additionally, we detect a modulation of this interaction by addition of recombinant Slit-2. Down-regulation of Robo-3 expression via small interfering RNA neutralizes Slit-2 induced migration block in PCI52_PC cells. In summary, our experiments show antitumorigenic effects of Slit-2 on P-cadherin expressing OSCC cells supposedly via modulation of Robo-3 interaction.


Subject(s)
Cadherins/metabolism , Carcinoma, Squamous Cell/metabolism , Cell Movement , Intercellular Signaling Peptides and Proteins/metabolism , Laryngeal Neoplasms/metabolism , Mouth Neoplasms/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Immunologic/metabolism , Blotting, Western , Cadherins/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Humans , Immunoenzyme Techniques , Immunoprecipitation , Intercellular Signaling Peptides and Proteins/genetics , Laryngeal Neoplasms/genetics , Laryngeal Neoplasms/pathology , Mouth Mucosa/metabolism , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Nerve Tissue Proteins/genetics , Phosphorylation , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Receptors, Cell Surface , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , beta Catenin/antagonists & inhibitors , beta Catenin/metabolism
10.
Pigment Cell Melanoma Res ; 24(1): 148-64, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20977688

ABSTRACT

A central event in the development of malignant melanoma is the loss of the tumor-suppressor protein E-cadherin. Here, we report that this loss is linked to the activation of the proto-oncogene c-Jun, a key player in tumorigenesis. In vivo, malignant melanomas show strong expression of the c-Jun protein in contrast to melanocytes. Interestingly, c-Jun mRNA levels did not differ in the melanoma cell lines when compared to melanocytes, suggesting that c-Jun could be regulated at the post-transcriptional level. To uncover the link between E-cadherin and c-Jun, we re-expressed E-cadherin in melanoma cells and detected decreased protein expression and activity of c-Jun. Furthermore, c-Jun accumulation is dependent on active E-cadherin-mediated cell-cell adhesion and regulated via the cytoskeleton. Additionally, we determined that, with respect to c-Jun regulation, there are two melanoma subgroups. One subgroup regulates c-Jun expression via the newly discovered E-cadherin-dependent signaling pathway, whereas the other subgroup uses the MAPKinases to regulate its expression. In summary, our data provide novel insights into the tumor-suppressor function of E-cadherin, which contributes to the suppression of c-Jun protein translation and transcriptional activity independent of MAPKinases.


Subject(s)
Cadherins/metabolism , Gene Expression Regulation, Neoplastic , Melanoma/genetics , Proto-Oncogene Proteins c-jun/genetics , Transcription, Genetic , Antigens, CD , Cell Adhesion , Cell Line, Tumor , Cytoskeleton/metabolism , Humans , Melanoma/enzymology , Melanoma/pathology , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins c-jun/metabolism
11.
Osteoarthritis Cartilage ; 19(2): 206-12, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21134476

ABSTRACT

OBJECTIVE: Recently, the transcription factor AP-2ɛ was shown to be a regulator of hypertrophy in cartilage and to be differentially expressed in osteoarthritis (OA). However, the only known target gene of AP-2ɛ up to date is integrin alpha10. To better characterize the function of AP-2ɛ in cartilage we screened for additional target genes. DESIGN: Promoter analysis, ChIP-assays and electrophoretic mobility shift assay were used to characterize the regulation of a new AP-2ɛ target gene in detail. RESULTS: In this study, we determined the chemokine CXCL1, already known to be important in osteoarthritis (OA), as a new target gene of AP-2ɛ. We could confirm that CXCL1 is expressed in chondrocytes and significantly over-expressed in OA-chondrocytes. Transient transfection of chondrocytes with an AP-2ɛ expression construct led to a significant increase of the CXCL1 mRNA level in these cells. We identified three potential AP-2 binding sites within the CXCL1 promoter and performed luciferase assays, indicating that an AP-2 binding motif (AP-2.2) ranging from position -135 to -144 bp relative to the translation start is responsive to AP-2ɛ. This result was further addressed by site-directed mutagenesis demonstrating that activation of the CXCL1 promoter by AP-2ɛ is exclusively dependent on AP-2.2. Chromatin immunoprecipitation and electromobility shift assays confirmed the direct binding of AP-2ɛ to the CXCL1 promoter in OA-chondrocytes at this site. CONCLUSION: These findings revealed CXCL1 as a novel target gene of AP-2ɛ in chondrocytes and support the important role of AP-2ɛ in cartilage.


Subject(s)
Cartilage/metabolism , Chemokine CXCL1/metabolism , Osteoarthritis/metabolism , Transcription Factor AP-2/metabolism , Aged , Cells, Cultured , Chondrocytes/metabolism , Electrophoresis , Female , Gene Expression Regulation , Humans , Male , Middle Aged , RNA, Messenger/metabolism , Transcription Factor AP-2/genetics , Transfection
13.
Cell Death Dis ; 1: e97, 2010 Nov 11.
Article in English | MEDLINE | ID: mdl-21368873

ABSTRACT

Melanoma inhibitory activity/cartilage-derived retinoic acid-sensitive protein (MIA/CD-RAP) is a small soluble protein secreted from chondrocytes. It was identified as the prototype of a family of extracellular proteins adopting an SH3 domain-like fold. In order to study the consequences of MIA/CD-RAP deficiency in detail we used mice with a targeted gene disruption of MIA/CD-RAP (MIA-/-) and analyzed cartilage organisation and differentiation in in vivo and in vitro models. Cartilage formation and regeneration was determined in models for osteoarthritis and fracture healing in vivo, in addition to in vitro studies using mesenchymal stem cells of MIA-/- mice. Interestingly, our data suggest enhanced chondrocytic regeneration in the MIA-/- mice, modulated by enhanced proliferation and delayed differentiation. Expression analysis of cartilage tissue derived from MIA-/- mice revealed strong downregulation of nuclear RNA-binding protein 54-kDa (p54(nrb)), a recently described modulator of Sox9 activity. In this study, we present p54(nrb) as a mediator of MIA/CD-RAP to promote chondrogenesis. Taken together, our data indicate that MIA/CD-RAP is required for differentiation in cartilage potentially by regulating signaling processes during differentiation.


Subject(s)
Cartilage/physiology , Extracellular Matrix Proteins/physiology , Regeneration , Animals , Cartilage/anatomy & histology , Cartilage/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Chondrogenesis , Disease Models, Animal , Down-Regulation , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/genetics , Mesenchymal Stem Cells/cytology , Mice , Mice, Knockout , Nuclear Matrix-Associated Proteins/metabolism , RNA-Binding Proteins/metabolism , SOX9 Transcription Factor/metabolism
14.
Int J Immunopathol Pharmacol ; 22(3): 715-22, 2009.
Article in English | MEDLINE | ID: mdl-19822088

ABSTRACT

Changes in the expression of repellent factors, i.e., Netrins and their receptors, may be responsible for the invasive behavior of the synovial tissue cells in patients with rheumatoid arthritis (RA) and osteoarthritis (OA). This study was carried out to analyze the expression of Netrins and their receptors in synovial cells of patients with RA, OA, and control subjects without synovial inflammation. Quantitative RT-PCR was performed to measure the expression of Netrin-1, -3, -4, Neogenin, DCC, UNC5A-D. The influence of Netrin-1 on synovial fibroblasts (SF) was analyzed by determining proliferation, migration, and their ability to organize collagen. SF expressed all repellent factors of the Netrin family. When comparing SF of healthy donors to patients with RA and OA, a stronger expression of UNC5B (4 fold) and UNC5C (769 fold) in RA and OA was found, whereas expression of the other molecules revealed no significant differences. Treating the SF-cells with recombinant Netrin-1 resulted in inhibition of migration of RA- and OA-SFs whereas control cells were not affected. The stronger expression of UNC5B and UNC5C receptors might contribute to the disordered phenotype of RA- and OA-SFs. Addition of Netrin-1 reduces the migratory ability of SFs, potentially by repulsion, as seen in neuronal cells in embryonic development. Due to its function, Netrin-1 may constitute a novel target in the treatment of OA and RA.


Subject(s)
Arthritis, Rheumatoid/metabolism , Cell Movement , Fibroblasts/metabolism , Nerve Growth Factors/metabolism , Osteoarthritis/metabolism , Receptors, Cell Surface/metabolism , Synovial Membrane/metabolism , Tumor Suppressor Proteins/metabolism , Adult , Aged , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Cartilage/metabolism , Case-Control Studies , Cells, Cultured , Collagen/metabolism , DCC Receptor , Female , Fibroblasts/pathology , Glycosaminoglycans/metabolism , Humans , Male , Membrane Proteins/genetics , Middle Aged , Nerve Growth Factors/genetics , Nerve Tissue Proteins/genetics , Netrin Receptors , Netrin-1 , Netrins , Osteoarthritis/genetics , Osteoarthritis/pathology , Receptors, Cell Surface/genetics , Recombinant Proteins/metabolism , Synovial Membrane/pathology , Tumor Suppressor Proteins/genetics
15.
Carcinogenesis ; 30(10): 1781-8, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19654099

ABSTRACT

Cadherins belong to a family of Ca(2+)-dependent homophilic cell-cell adhesion proteins that are important for correct cellular localization and tissue integrity. They play a major role in the development and homeostasis of epithelial architecture. Recently, it has become more and more evident that P-cadherin contributes to the oncogenesis of many tumors. To analyze the role of P-cadherin in oral squamous cell carcinoma (OSCC), we used a cell line that was deficient of the classical cadherins, P-cadherin, E-cadherin and N-cadherin. This cell line was transfected with full-length P-cadherin (PCI52_PC). After overexpression of P-cadherin, PCI52_PC gained an epithelial-like brickstone morphology in contrast to the mock-transfected cells with a spindle-shaped mesenchymal morphology. Immunohistochemical analysis revealed a strong nuclear Snail staining in mock-transfected cells compared with a significantly reduced nuclear staining and translocation to the cytoplasm in P-cadherin-overexpressing cells. Interestingly, the effects triggered by P-cadherin overexpression could be reversed by transfecting the cells with an antisense P-cadherin plasmid construct. Additional investigations showed a reexpression of E-cadherin in all P-cadherin-transfected cell clones in contrast to the mock controls. Analyzing the signaling mechanism behind it, we found glycogen-synthase-kinase-3beta (GSK-3beta) bound to Snail in all cell clones. Furthermore, P-cadherin-overexpressing cell lines showed activated GSK-3beta that phosphorylated Snail leading to its cytoplasmic translocation. In summary, our results reveal P-cadherin as one major component in reconfiguring mesenchymal cells with epithelial features by triggering GSK-3beta-mediated inactivation and cytoplasmatic translocation of Snail in OSCC.


Subject(s)
Cadherins/toxicity , Carcinoma, Squamous Cell/chemically induced , Glycogen Synthase Kinase 3/metabolism , Mouth Neoplasms/chemically induced , Transcription Factors/metabolism , Cadherins/deficiency , Cadherins/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement , DNA, Neoplasm/genetics , DNA, Neoplasm/isolation & purification , Gene Amplification , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , HeLa Cells , Humans , Immunohistochemistry , Kinetics , Mesoderm/drug effects , Mesoderm/physiology , Mouth Neoplasms/pathology , Neoplasm Invasiveness , Promoter Regions, Genetic , Snail Family Transcription Factors , Transfection
16.
Br J Cancer ; 101(4): 551-6, 2009 Aug 18.
Article in English | MEDLINE | ID: mdl-19638982

ABSTRACT

Analysis of microRNA (miRNA) biogenesis and function is an area of research that started only recently but has subsequently accelerated tremendously. This is because of the impressive impact of miRNA-mediated gene regulation and the obvious potential of those tiny RNA molecules in future diagnostic and therapeutic applications. In this review, recent progress to reveal the role of miRNAs in the tumourigenesis of malignant melanoma, as well as future prospects of melanoma-related miRNA research, will be addressed.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Melanoma/genetics , MicroRNAs/genetics , Skin Neoplasms/genetics , Animals , Gene Expression Profiling , Humans
17.
Oncogene ; 27(52): 6698-706, 2008 Nov 06.
Article in English | MEDLINE | ID: mdl-18679415

ABSTRACT

Although integrin beta(3) is known to play an important role in melanoma progression and invasion, regulation of integrin beta(3) expression in melanoma has not been analysed in detail until today. As transcriptional regulation of integrin beta(3) was ruled out by our analysis, we concentrated on the regulation by microRNAs (miRNAs). Comparing primary melanocytes and malignant melanoma cell lines, we found that one candidate miRNA, miR-let-7a, was lost in melanoma and sequence analysis suggested an interaction with the 3'-untranslated region (3'-UTR) of integrin beta(3) mRNA. Transfection of melanoma cells with let-7a pre-miR(TM) molecules resulted in the downregulation of integrin beta(3) mRNA and protein expression. In addition, we cloned the 3'-UTR of the integrin beta(3) mRNA containing the let-7a target sequence into a reporter plasmid and revealed that let-7a negatively regulates reporter gene expression. The repressed expression of integrin beta(3) accompanies with reduced invasive potential of melanoma cells transfected with synthetic let-7a molecules observed in Boyden chamber assays. On the other hand, the induction of integrin beta(3) expression was achieved in melanocytes by transfection with let-7a anti-miRs, resulting in invasive behavior of transfected melanocytes. In summary, we determined miRNA let-7a to be an important regulator of integrin beta(3) expression and showed that the loss of let-7a expression is involved in development and progression of malignant melanoma.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Integrin beta3/genetics , Integrin beta3/metabolism , Melanoma/genetics , Melanoma/metabolism , MicroRNAs/genetics , Cell Line
18.
FEBS J ; 275(16): 4198-210, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18637117

ABSTRACT

Cadherins belong to a family of homophilic cell-cell adhesion proteins that are responsible for the establishment of a precise cell architecture and tissue integrity. Moreover, experimental data suggest that loss of intercellular adhesion is inversely correlated with cellular differentiation. Furthermore, dedifferentiation is closely linked to tumor progression. Recently, we have shown that a secreted 50 kDa N-terminal fragment of P-cadherin plays a role in the progression of malignant melanoma. In this study, we have detected both the full-length and the truncated versions of P-cadherin in cell lysates of differentiated head and neck oral squamous cell carcinoma cell lines, whereas in cell lysates of dedifferentiated cell lines, we detected only the truncated 50 kDa version of P-cadherin. Treatment of the cell lines with a recombinantly expressed biotinylated, soluble 50 kDa form of the N-terminal part of P-cadherin revealed a major effect on cell aggregation and migration of oral squamous cell carcinoma cells. However, the 50 kDa N-terminal fragment of P-cadherin did not show any influence on cell proliferation in 2D and 3D cell culture. These results suggest that generation of truncated P-cadherin during the progression of oral squamous carcinoma attenuates tissue integrity, facilitates cellular separation, and leads to the acquisition of a more migratory phenotype.


Subject(s)
Cadherins/metabolism , Carcinoma, Squamous Cell/metabolism , Mouth Neoplasms/metabolism , Cadherins/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Differentiation , Cell Line, Tumor , Cell Movement , Humans , Male , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Sequence Deletion
19.
Histol Histopathol ; 23(4): 391-5, 2008 04.
Article in English | MEDLINE | ID: mdl-18228195

ABSTRACT

In general, it is believed that the extraction of proteins from formalin-fixed paraffin embedded samples is not feasible. However, recently a new technique was developed, presenting the extraction of non-degraded, full length proteins from formalin fixed tissues, usable for western blotting and protein arrays. In the study presented here, we applied this technique to punch biopsies of formalin fixed tissues embedded in paraffin to reduce heterogeneity of the tissue represented in sections, and to ensure analysing mainly defined cellular material. Successful extraction was achieved even from very small samples (0.7 mm(3)). Additionally, we were able to detect highly glycosylated proteins and protein modification, such as phosphorylation. Interestingly, with this technique it is feasible to extract high quality proteins from 14 year old samples. In summary, the new technique makes a great pool of material now usable for molecular analysis with high throughput tools.


Subject(s)
Fixatives/chemistry , Formaldehyde/chemistry , Proteins/isolation & purification , Antigens, Neoplasm , Biopsy , Blotting, Western , Cadherins/analysis , Cadherins/isolation & purification , Electrophoresis, Polyacrylamide Gel , Feasibility Studies , Freezing , Humans , Liver/chemistry , Liver/surgery , Melanoma/chemistry , Melanoma/surgery , Models, Biological , Neoplasm Proteins , Paraffin Embedding , Silver Staining , Tissue Fixation , Tumor Suppressor Proteins/analysis , Tumor Suppressor Proteins/isolation & purification
20.
Gut ; 57(2): 243-51, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17881540

ABSTRACT

BACKGROUND: Melanoma inhibitory activity 2 (MIA2) is a novel gene of the MIA gene family. The selective expression of MIA2 in hepatocytes is controlled by hepatocyte nuclear factor (HNF) 1 binding sites in the MIA2 promotor. In contrast, in most hepatocellular carcinomas (HCC) MIA2 expression is down-regulated or lost. AIM: In this study we examined the regulation and functional role of MIA2 in hepatocancerogenesis. METHODS AND RESULTS: In HCC cell lines and tissues HNF-1 expression was lower than in primary human hepatocytes (PHH) and corresponding non-tumorous tissue, respectively, and correlated significantly with the down-regulation of MIA2 expression. Re-expression of HNF-1 in HCC cells reinduced MIA2 in HCC cells to similar levels as found in PHH. Further, MIA2 was re-expressed in HCC cell lines by stable transfection, and the generated cell clones revealed a strongly reduced invasive potential and proliferation rate in vitro. In line with these findings treatment of HCC cells with recombinant MIA2 inhibited proliferation and invasion. In nude mice MIA2 re-expressing HCC cells grew significantly slower and revealed a less invasive growth pattern. Immunohistochemical analysis of a tissue microarray containing HCC and corresponding non-cancerous liver tissue of 85 patients confirmed reduced MIA2 expression in HCC. Furthermore, MIA2 negative HCC tissue showed a significantly higher Ki67 labelling index and loss of MIA2 expression correlated significantly with more advanced tumour stages. CONCLUSION: This study presents MIA2 as an inhibitor of HCC growth and invasion both in vitro and in vivo, and consequently, as a tumour suppressor of HCC. Further, our findings indicate a novel mechanism, how loss of HNF-1 expression in HCC affects tumorigenicity via down-regulation of MIA2.


Subject(s)
Carcinoma, Hepatocellular/genetics , Extracellular Matrix Proteins/genetics , Genes, Tumor Suppressor , Liver Neoplasms/genetics , Neoplasm Proteins/genetics , Tumor Suppressor Proteins/genetics , Adult , Aged , Aged, 80 and over , Antigens, Neoplasm , Cell Cycle , Cell Proliferation , Cells, Cultured , Down-Regulation , Extracellular Matrix Proteins/antagonists & inhibitors , Female , Hepatocyte Nuclear Factor 1/metabolism , Humans , Male , Middle Aged , Neoplasm Proteins/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...