Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
PLoS Biol ; 16(9): e2006624, 2018 09.
Article in English | MEDLINE | ID: mdl-30261040

ABSTRACT

Head and neck squamous cell carcinomas (HNSCCs) are characterized by outstanding molecular heterogeneity that results in severe therapy resistance and poor clinical outcome. Inter- and intratumoral heterogeneity in epithelial-mesenchymal transition (EMT) was recently revealed as a major parameter of poor clinical outcome. Here, we addressed the expression and function of the therapeutic target epidermal growth factor receptor (EGFR) and of the major determinant of epithelial differentiation epithelial cell adhesion molecule (EpCAM) in clinical samples and in vitro models of HNSCCs. We describe improved survival of EGFRlow/EpCAMhigh HNSCC patients (n = 180) and provide a molecular basis for the observed disparities in clinical outcome. EGF/EGFR have concentration-dependent dual capacities as inducers of proliferation and EMT through differential activation of the central molecular switch phosphorylated extracellular signal-regulated kinase 1/2 (pERK1/2) and EMT transcription factors (EMT-TFs) Snail, zinc finger E-box-binding homeobox 1 (Zeb1), and Slug. Furthermore, soluble ectodomain of EpCAM (EpEX) was identified as a ligand of EGFR that activates pERK1/2 and phosphorylated AKT (pAKT) and induces EGFR-dependent proliferation but represses EGF-mediated EMT, Snail, Zeb1, and Slug activation and cell migration. EMT repression by EpEX is realized through competitive modulation of pERK1/2 activation strength and inhibition of EMT-TFs, which is reflected in levels of pERK1/2 and its target Slug in clinical samples. Accordingly, high expression of pERK1/2 and/or Slug predicted poor outcome of HNSCCs. Hence, EpEX is a ligand of EGFR that induces proliferation but counteracts EMT mediated by the EGF/EGFR/pERK1/2 axis. Therefore, the emerging EGFR/EpCAM molecular cross talk represents a promising target to improve patient-tailored adjuvant treatment of HNSCCs.


Subject(s)
Epidermal Growth Factor/metabolism , Epithelial Cell Adhesion Molecule/chemistry , Epithelial-Mesenchymal Transition , Extracellular Signal-Regulated MAP Kinases/metabolism , Head and Neck Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Enzyme Activation/drug effects , ErbB Receptors/chemistry , ErbB Receptors/metabolism , Head and Neck Neoplasms/pathology , Humans , Ligands , Models, Biological , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Domains , Proto-Oncogene Proteins c-akt/metabolism , Snail Family Transcription Factors/metabolism , Treatment Outcome
2.
J Biol Chem ; 293(23): 8994-9005, 2018 06 08.
Article in English | MEDLINE | ID: mdl-29700109

ABSTRACT

Epithelial cell-adhesion molecule (EpCAM) is a transmembrane protein that regulates cell cycle progression and differentiation and is overexpressed in many carcinomas. The EpCAM-induced mitogenic cascade is activated via regulated intramembrane proteolysis (RIP) of EpCAM by ADAM and γ-secretases, generating the signaling-active intracellular domain EpICD. Because of its expression pattern and molecular function, EpCAM is a valuable target in prognostic and therapeutic approaches for various carcinomas. So far, several immunotherapeutic strategies have targeted the extracellular domain of EpCAM. However, targeting the intracellular signaling cascade of EpCAM holds promise for specifically interfering with EpCAM's proliferation-stimulating signaling cascade. Here, using a yellow fluorescence protein-tagged version of the C-terminal fragment of EpCAM, we established a high-content screening (HCS) of a small-molecule compound library (n = 27,280) and characterized validated hits that target EpCAM signaling. In total, 128 potential inhibitors were initially identified, of which one compound with robust inhibitory effects on RIP of EpCAM was analyzed in greater detail. In summary, our study demonstrates that the development of an HCS for small-molecule inhibitors of the EpCAM signaling pathway is feasible. We propose that this approach may also be useful for identifying chemical compounds targeting other disorders involving membrane cleavage-dependent signaling pathways.


Subject(s)
Epithelial Cell Adhesion Molecule/antagonists & inhibitors , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology , Cell Proliferation/drug effects , Drug Evaluation, Preclinical/methods , Epithelial Cell Adhesion Molecule/metabolism , HEK293 Cells , High-Throughput Screening Assays/methods , Humans , Small Molecule Libraries/chemistry , Transcription, Genetic/drug effects
3.
J Biol Chem ; 290(40): 24574-91, 2015 Oct 02.
Article in English | MEDLINE | ID: mdl-26292218

ABSTRACT

Human epithelial cell adhesion molecule (HEPCAM) is a tumor-associated antigen frequently expressed in carcinomas, which promotes proliferation after regulated intramembrane proteolysis. Here, we describe extracellular shedding of HEPCAM at two α-sites through a disintegrin and metalloprotease (ADAM) and at one ß-site through BACE1. Transmembrane cleavage by γ-secretase occurs at three γ-sites to generate extracellular Aß-like fragments and at two ϵ-sites to release human EPCAM intracellular domain HEPICD, which is efficiently degraded by the proteasome. Mapping of cleavage sites onto three-dimensional structures of HEPEX cis-dimer predicted conditional availability of α- and ß-sites. Endocytosis of HEPCAM warrants acidification in cytoplasmic vesicles to dissociate protein cis-dimers required for cleavage by BACE1 at low pH values. Intramembrane cleavage sites are accessible and not part of the structurally important transmembrane helix dimer crossing region. Surprisingly, neither chemical inhibition of cleavage nor cellular knock-out of HEPCAM using CRISPR-Cas9 technology impacted the adhesion of carcinoma cell lines. Hence, a direct function of HEPCAM as an adhesion molecule in carcinoma cells is not supported and appears to be questionable.


Subject(s)
Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/metabolism , Amino Acids/chemistry , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Binding Sites , Cell Adhesion , Cell Line, Tumor , Cell Membrane/metabolism , Cell Separation , Endocytosis , Epithelial Cell Adhesion Molecule , Flow Cytometry , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Protein Multimerization , Protein Structure, Tertiary , Proteolysis
5.
Hepatol Int ; 10(2): 370-6, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26646663

ABSTRACT

BACKGROUND/PURPOSE OF THE STUDY: Despite tremendous progress in therapy, about 30% of patients with hepatoblastoma still succumb to the disease. Thus, the development of improved therapies as well as the identification of prognostic factors are urgently needed. METHODS: In the present study, expression and promoter methylation of the N-myc downstream-regulated gene (NDRG2), a tumor suppressor gene contributing to the regulation of the Wnt signalling pathway, was analysed in 38 hepatoblastoma samples by real-time reverse transcription-PCR and pyrosequencing, respectively. RESULTS: The NDRG2 gene was highly expressed in normal pediatric liver tissue, but was significantly downregulated in heptoblastoma primary tumors. Detailed methylation analysis of CpG sites in the NDRG2 promoter region revealed a general high degree of DNA methylation in hepatoblastoma, which correlated with the suppression of NDRG2. By analyzing clinicopathological features we could demonstrate a strong association between low NDRG2 expression and tumor metastasis. Importantly, the overall survival analysis by Kaplan-Meier revealed that high NDRG2 expression was correlated with a higher survival rate in hepatoblastoma patients. CONCLUSION: Our data show that downregulation of NDRG2 may play an important role in advanced hepatoblastomas.


Subject(s)
Down-Regulation , Gene Expression Regulation, Neoplastic , Hepatoblastoma/genetics , Liver Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Child , Child, Preschool , DNA Methylation , Female , Hepatoblastoma/pathology , Humans , Infant , Liver Neoplasms/pathology , Male , Neoplasm Metastasis , Prognosis , Promoter Regions, Genetic , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL