Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Recept Signal Transduct Res ; 37(1): 25-37, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27051967

ABSTRACT

INTRODUCTION: Receptors of the ErbB family belong to the key players in cancer development and are targets of several therapeutic approaches. Their functional dependency on the tumor microenvironment, especially on CAFs is albeit still poorly understood. Our objective was to investigate the impact of CAF secretome on ErbB receptor expression and signaling behavior in OSCC. METHODS: Stimulation of PE/CA-PJ15 OSCC cells with conditioned media of TGF-ß1-activated fibroblasts was used as model system for CAF to cancer cell communication. Thereby costimulation with inhibitors against matrix metalloproteinases (MMPs), epidermal growth factor receptor (EGFR), MAPK/ERK kinase (MEK), phosphoinositide-3 kinase (PI3-K), signal transducer and activator of transcription 3 (Stat3) or knockdown of Her3 by siRNA was utilized for detailed investigation of the expression, dimerization and signaling pattern of ErbB in western blot and coimmunoprecipitation. RESULTS: Our results show that soluble factors in activated fibroblast secretome stimulate metalloproteinase activity in the membrane of cancer cells. Thereby ligands are released that activate EGFR and subsequently upregulates EGFR expression via the STAT3 pathway. Simultaneously, the expression of PKCɛ was enhanced via a PI3-kinase/Akt-mediated pathway and a negative feedback regulation loop on EGFR downstream signaling generated. Furthermore, the activated fibroblasts secretome stimulated the highly oncogenic hetero-dimerization between HER3 and p95HER2. That protein association is inversely dependent on the expression level of HER3. CONCLUSIONS: Our results demonstrate that the activated fibroblasts secretome can induce a counterbalanced regulation of protein expression, downstream signaling and the dimerization patterns of different ErbB receptor subtypes in the cancer cell. Thus, the combinatorial targeting of CAFs and selective ErbB receptor subtype inhibitors may provide a useful approach in cancer therapy.


Subject(s)
Carcinoma, Squamous Cell/pathology , Gene Expression Regulation , Mouth Neoplasms/pathology , Myofibroblasts/pathology , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Signal Transduction , Blotting, Western , Carcinoma, Squamous Cell/metabolism , Cell Cycle , Cell Proliferation , Cells, Cultured , ErbB Receptors/metabolism , Humans , Immunoprecipitation , Mouth Neoplasms/metabolism , Myofibroblasts/metabolism , Protein Multimerization , Receptor, ErbB-2/chemistry , Receptor, ErbB-3/chemistry
2.
Exp Cell Res ; 322(2): 402-14, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24394543

ABSTRACT

Crosstalk between carcinoma associated fibroblasts (CAFs) and oral squamous cell carcinoma (OSCC) cells is suggested to mediate phenotype transition of cancer cells as a prerequisite for tumour progression, to predict patients' outcome, and to influence the efficacy of EGFR inhibitor therapies. Here we investigate the influence of activated fibroblasts as a model for CAFs on phenotype and EGFR signalling in OSCC cells in vitro. For this, immortalised hTERT-BJ1 fibroblasts were activated with TGFß1 and PDGFAB to generate a myofibroblast or proliferative phenotype, respectively. Conditioned media (FCMTGF, FCMPDGF) were used to stimulate PE/CA-PJ15 OSCC cells. Results were compared to the effect of conditioned media of non-stimulated fibroblasts (FCMB). FCMTGF stimulation leads to an up-regulation of vimentin in the OSCC cells and an enhancement of invasive behaviour, indicating EMT-like effects. Similarly, FCMTGF≫FCMPDGF induced up-regulation of EGFR, but not of ErbB2/ErbB3. In addition, we detected an increase in basal activities of ERK, PI3K/Akt and Stat3 (FCMTGF>FCMPDGF) accompanied by protein interaction of vimentin with pERK. These effects are correlated with an increased proliferation. In summary, our results suggest that the activated myofibroblast phenotype provides soluble factors which are able to induce EMT-like phenomena and to increase EGFR signalling as well as cell proliferation in OSCC cells. Our results indicate a possible influence of activated myofibroblasts on EGFR-inhibitor therapy. Therefore, CAFs may serve as promising novel targets for combined therapy strategies.


Subject(s)
Carcinoma, Squamous Cell/pathology , Cell Cycle/physiology , Epithelial-Mesenchymal Transition , ErbB Receptors/metabolism , Fibroblasts/pathology , Mouth Neoplasms/pathology , Myofibroblasts/pathology , Blotting, Western , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Culture Media, Conditioned/pharmacology , ErbB Receptors/genetics , Fibroblasts/metabolism , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Immunoprecipitation , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Mutation/genetics , Myofibroblasts/metabolism , Neoplasm Invasiveness , Phenotype , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Tumor Cells, Cultured , Vimentin/genetics , Vimentin/metabolism
3.
Oncotarget ; 3(1): 31-43, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22289787

ABSTRACT

Head and neck squamous cell carcinomas (HNSCCs) are the sixth most common malignant neoplasm and more than 50% of patients succumb to this disease. HNSCCs are characterized by therapy resistance, which relies on the overexpression of anti-apoptotic proteins and on the aberrant regulation of the epidermal growth factor receptor (EGFR). As inherent and acquired resistance to therapy counteracts improvement of long-term survival, novel multi-targeting strategies triggering cancer cell death are urgently required. We investigated how induction of replicational stress by the ribonucleotide reductase inhibitor hydroxyurea (HU) combined with histone deacetylase inhibitors (HDACi) exerts anti-tumor activity. We treated HNSCC cell lines and freshly isolated tumor cells with HDACi, such as the clinically approved anti-epileptic drug valproic acid (VPA), in combination with HU. Our data demonstrate that at clinically achievable levels VPA/HU combinations efficiently block proliferation as well as clonogenic survival, and trigger apoptosis of HNSCC cells. In the presence of VPA/HU, such tumor cells increase expression of the pro-apoptotic BCL-2 family protein BIM, independent of wild-type p53 signaling and in the absence of increased expression of the p53 targets PUMA and BAX. The pro-apoptotic activity of BIM in HNSCCs was found critical for tumor cell death; ectopic overexpression of BIM induced HNSCC apoptosis and RNAi-mediated depletion of BIM protected HNSCC cells from VPA/HU. Also, significantly elevated BIM levels (p less than 0.01) were detectable in the apoptotic tumor centers versus proliferating tumor margins in HNSCC patients (n=31), underlining BIM's clinical relevance. Importantly, VPA/HU treatment additionally reduces expression and cell surface localization of EGFR. Accordingly, in a xenograft mouse model, VPA/HU efficiently blocked tumor growth (P less than 0.001) correlating with BIM induction and EGFR downregulation. We provide a molecular rationale for the potent anti-cancer activities of this drug combination. Our data suggest its exploitation as a potential strategy for the treatment of HNSCC and other tumor entities characterized by therapy resistance linked to dysregulated EGFR activation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis Regulatory Proteins/physiology , Apoptosis/drug effects , Carcinoma, Squamous Cell/drug therapy , Enzyme Inhibitors/administration & dosage , Genes, erbB-1/drug effects , Head and Neck Neoplasms/drug therapy , Membrane Proteins/physiology , Proto-Oncogene Proteins/physiology , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Down-Regulation/drug effects , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Head and Neck Neoplasms/pathology , Histone Deacetylase Inhibitors/administration & dosage , Histone Deacetylase Inhibitors/pharmacology , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Nude , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Ribonucleotide Reductases/antagonists & inhibitors , Squamous Cell Carcinoma of Head and Neck , Xenograft Model Antitumor Assays
4.
Cell Signal ; 24(2): 422-434, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21964064

ABSTRACT

Recently, we described a novel function of over-expressed protein kinase Cε (PKCε) as a negative allosteric modulator of EGFR signalling in several head and neck squamous carcinoma (HNSCC) cell lines. Extending this work, here we present several lines of evidence for the potency of PKCε to differently modulate the efficacy of EGFR tyrosine kinase inhibitors (TKIs) such as gefitinib and lapatinib. Using the HNSCC cell line FaDu as a model, we demonstrate by co-immunoprecipitation the physical association of over-expressed PKCε with the EGFR which is stabilised by gefitinib and leads to an increase in gefitinib-induced inhibition of EGFR downstream signalling and elevated EGFR-ErbB2 heterodimerisation. Cell cycle and Western blot analysis revealed that the gefitinib-induced apoptosis was enhanced whereas the pro-apoptotic effect of lapatinib that requires another EGFR conformation was reduced by PKCε. Our findings suggest that due to elevated expression PKCε may associate with the EGFR resulting in conformational changes and different allosteric modulation of the EGFR behaviour towards TKIs. This surprising capacity indicates PKCε as a novel predictive marker protein in molecular cancer therapy with EGFR tyrosine kinase inhibitors.


Subject(s)
Carcinoma, Squamous Cell/metabolism , ErbB Receptors/metabolism , Head and Neck Neoplasms/metabolism , Protein Kinase C-epsilon/metabolism , Quinazolines/pharmacology , Receptor, ErbB-2/metabolism , Signal Transduction/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Blotting, Western , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Dimerization , ErbB Receptors/chemistry , ErbB Receptors/genetics , Flow Cytometry , Gefitinib , Gene Expression , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , Immunoprecipitation , Lapatinib , Plasmids , Protein Conformation , Protein Kinase C-epsilon/antagonists & inhibitors , Protein Kinase C-epsilon/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Quinazolines/therapeutic use , Receptor, ErbB-2/genetics , Signal Transduction/genetics , Transfection
5.
Cell Signal ; 23(2): 436-48, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20965248

ABSTRACT

Protein kinase C ε (PKCε) is a transforming oncogene and plays a pivotal role in numerous cellular processes including proliferation, invasion and differentiation. Recently, we described a function of PKCε as a scaffold protein linking PLCγ1 to the EGFR module. Here, in the head and neck squamous carcinoma cell line (HNSCC) FaDu we demonstrate that over-expressed PKCε may be associated with the EGFR. This is linked with the consecutive inhibition of the recruitment of PLCγ1 to the EGFR, of the catalytical activation of PLCγ1 by EGF, and of the PLCγ1-mediated effect of EGF on cell proliferation. These effects are independent of the catalytical as well as the scaffold activity of PKCε but are a function of the cellular expression level of PKCε. In contrast to FaDu cells where the PLCγ1 pathway was selectively affected, in three other HNSCC cell lines investigated over-expression of PKCε resulted in association with EGFR and, subsequently, in either partial (ERK and Akt or PLCγ1 and Akt) or complete (ERK, PLCγ1 and Akt) inhibition of the main EGFR signalling pathways. Together, our data suggest that in particular carcinoma cells highly expressed PKCε may act as negative allosteric modulator of EGFR signalling. This novel function of PKCε provides also the first indication that the EGFR may be a target for allosteric modulation by accessory proteins.


Subject(s)
ErbB Receptors/physiology , Protein Kinase C-epsilon/physiology , Allosteric Regulation , Animals , COS Cells , Cell Movement , Cell Proliferation , Chlorocebus aethiops , Humans , Mice , Phospholipase C gamma/metabolism , Signal Transduction , Tumor Cells, Cultured
6.
Mol Cell Endocrinol ; 331(2): 222-31, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-20398727

ABSTRACT

About one decade ago has been demonstrated that G protein-coupled receptors (GPCRs) are able to utilize the epidermal growth factor (EGF) receptor (EGFR) as signalling intermediate. Thereby GPCRs are enabled to regulate cell growth, differentiation, and migration. A molecular mechanism for this process has been proposed that involves the activation of a distinct set of metalloproteases and the subsequent generation and release of particular members of the EGF peptide family which in turn activate the EGFR in an autocrine/paracrine manner. This model that allows GPCRs direct access to the signalling network of the EGFR family has emerged as a valid concept in a variety of cell types including cancer cells. The present review briefly summarizes the current knowledge but will be focussed on the ligand-dependency of EGFR transactivation. Several alternative mechanisms and novel aspects will be introduced. Using the example of head and neck squamous carcinoma, the potency of EGFR transactivation as a therapeutical target will be discussed.


Subject(s)
ErbB Receptors/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Carcinoma/metabolism , Carcinoma/therapy , Carcinoma, Squamous Cell , Cell Growth Processes/physiology , Cell Movement/physiology , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/therapy , Humans , Ligands , Metalloproteases/metabolism , Neoplasms, Squamous Cell/metabolism , Neoplasms, Squamous Cell/therapy , Signal Transduction , Squamous Cell Carcinoma of Head and Neck
7.
Mol Cancer Res ; 8(5): 783-97, 2010 May.
Article in English | MEDLINE | ID: mdl-20407013

ABSTRACT

The atypical protein kinase Czeta (PKCzeta) was recently shown to mediate epidermal growth factor (EGF)-induced activation of extracellular signal-regulated kinase (ERK) in head and neck squamous carcinoma (HNSCC) cells. Here, it is shown that EGF may induce tyrosine phosphorylation of PKCzeta in several HNSCC cells, breast carcinoma cells, as well as mouse embryonic fibroblasts. In COS-7 cells overexpressing EGF receptor (EGFR) and PKCzeta as a tumor cell model, we show that PKCzeta tyrosine phosphorylation by EGF is induced by catalytic activation. Using a loss-of-function mutant of PKCzeta, we can show that the tyrosine residue 417 in PKCzeta plays an important role in both PKCzeta activation and the ability of PKCzeta to mediate activation of ERK. The importance of PKCzeta in EGF-induced ERK activation can also be shown in several HNSCC and breast carcinoma cell lines as well as in PKCzeta-deficient mouse embryonic fibroblasts. In addition, we present several lines of evidence suggesting the physical association of PKCzeta with EGFR and the importance of the EGFR tyrosine kinase c-Src and the Src-specific phosphorylation site pY845-EGFR in the tyrosine phosphorylation as well as catalytic activation of PKCzeta. This study characterizes PKCzeta as a novel mitogenic downstream mediator of EGFR and indicates PKCzeta as a therapeutic target in some carcinomas.


Subject(s)
ErbB Receptors/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Protein Kinase C/metabolism , Signal Transduction/physiology , Tyrosine/metabolism , Animals , COS Cells , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/pathology , Catalytic Domain/genetics , Cell Line, Tumor , Chlorocebus aethiops , Enzyme Activation/genetics , Enzyme Activation/physiology , ErbB Receptors/genetics , Head and Neck Neoplasms/enzymology , Head and Neck Neoplasms/pathology , Humans , MAP Kinase Signaling System/physiology , Mice , Mitogen-Activated Protein Kinase 3/genetics , Phosphorylation/genetics , Protein Kinase C/genetics , Signal Transduction/genetics
8.
Cell Signal ; 19(9): 1830-43, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17561374

ABSTRACT

Phospholipase Cgamma1 (PLCgamma1) represents a major downstream signalling component of the epidermal growth factor (EGF) receptor (EGFR) and is activated by tyrosine phosphorylation. Here we show for the first time that cellular knockdown of protein kinase Cepsilon (PKCepsilon) leads to decreased activation of PLCgamma1 by EGF and that EGF induces tyrosine phosphorylation of PKCepsilon as well as association of PKCepsilon with both EGFR and PLCgamma1. Using several mutants, co-immunoprecipitation and phosphopeptide-based pull-down experiments we found that in dependency on c-Src and EGF-stimulation PKCepsilon may bind to the c-Src-specific phosphorylation site pY845-EGFR. Furthermore, we identified a single tyrosine residue, PKCepsilon-Y573, within a consensus binding sequence of the C-terminal SH2 domain of PLCgamma1 which is critical for both tyrosine phosphorylation of PKCepsilon and its association with PLCgamma1. Thus, in particular cells and independent of the kinase activity PKCepsilon may form a signalling module with EGFR and PLCgamma1. Thereby the tyrosine phosphorylation of PLCgamma1 via the EGFR may be facilitated. This is a novel function of PKCepsilon upstream of PLCgamma1 and a novel paradigm for the EGF-induced formation of multi-protein complexes.


Subject(s)
Epidermal Growth Factor/pharmacology , Phospholipase C gamma/metabolism , Protein Kinase C-epsilon/metabolism , Amino Acid Sequence , Animals , COS Cells , Catalysis/drug effects , Chlorocebus aethiops , Consensus Sequence , Enzyme Activation/drug effects , Enzyme Induction/drug effects , ErbB Receptors/metabolism , Humans , Immunoprecipitation , Molecular Sequence Data , Mutant Proteins/metabolism , Phospholipase C gamma/chemistry , Phosphotyrosine/metabolism , Protein Binding/drug effects , Protein Kinase C-epsilon/biosynthesis , Protein Kinase C-epsilon/chemistry , Proto-Oncogene Proteins pp60(c-src)/metabolism , RNA Interference , src Homology Domains
9.
Cell Signal ; 18(10): 1633-46, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16495036

ABSTRACT

Transactivation of epidermal growth factor receptor (EGFR) by G protein-coupled receptors (GPCRs) is currently understood to be mediated by matrix metalloproteases (MMPs) and the release of EGF-like ligands. This ligand-mediated process also suggests that downstream of EGFR the signalling in response to GPCR ligands or EGF appears to be indistinguishable. Here we provide evidence that transactivation of EGFR by the beta2-adrenergic receptor (beta2-AR) is independent of MMPs and results in an incomplete downstream signalling involving extracellular signal-activated kinase (ERK) but not PLCgamma1 and Akt. In contrast, beta2-AR has the ability to activate PLCgamma1 when the EGFR is primed either by co-stimulation with EGF or by increased basal activity due to over-expression. In that way but not via the beta2-AR-mediated transactivation the EGFR docking sites pY992 and pY1173 may be generated which are critical for PLCgamma1. This EGFR-supported transactivation is strongly dependent on EGFR tyrosine kinase, c-Src, and the c-Src-specific EGFR tyrosine residue 845 and represents a novel paradigm of EGFR transactivation.


Subject(s)
ErbB Receptors/metabolism , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction , Transcriptional Activation , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression , Humans , Ligands , Matrix Metalloproteinases/metabolism , Neoplasms/enzymology , Phospholipase C gamma/metabolism , Phosphorylation , Phosphotyrosine/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism
10.
Cell Signal ; 18(7): 1031-40, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16226010

ABSTRACT

In the COS-7 cell signalling network high levels of cAMP produced, for example, by co-stimulation of beta2-adrenergic receptor (beta2-AR) and bradykinin B2 receptor (BKR) may affect epidermal growth factor receptor (EGFR)-mediated activation of extracellular signal-stimulated kinase (ERK). In contrast, co-stimulation of either beta2-AR or B2R with EGFR leads to synergistic activation of ERK. Due to triple stimulation of these receptors the synergistic effects on ERK activation as well as cAMP accumulation are diminished. Here we demonstrate that EGF is capable of inducing Src-mediated phosphorylation of the tyrosine residues 177 and 347 of BKR. Their replacement by phenylalanine led to BKR mutants which are unable to activate the cAMP pathway. Using these mutants we can show that EGF attenuates but does not completely inhibit the BKR/cAMP pathway which is counteracting the EGFR signalling to ERK. Our findings suggest that the EGFR may control the cellular network rather by balancing mechanisms then by switch on/off reactions.


Subject(s)
ErbB Receptors/physiology , Extracellular Signal-Regulated MAP Kinases/physiology , Signal Transduction , Amino Acid Substitution , Animals , COS Cells , Chlorocebus aethiops , Cyclic AMP/physiology , Enzyme Activation , Epidermal Growth Factor/physiology , ErbB Receptors/agonists , Humans , Mutation , Phenylalanine/genetics , Phenylalanine/metabolism , Phosphorylation , Receptor, Bradykinin B2/genetics , Receptor, Bradykinin B2/physiology , Receptors, Adrenergic, beta-2/metabolism , Tyrosine/genetics , Tyrosine/metabolism , src-Family Kinases/physiology
11.
Cell Signal ; 18(8): 1338-49, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16337776

ABSTRACT

Transactivation of epidermal growth factor receptor (EGFR) by G protein-coupled receptors (GPCRs) has been attributed to the activation of matrix metalloproteases (MMPs) and the release of EGF family ligands such as HB-EGF. This mode of transactivation leads to signalling downstream of EGFR which is indistinguishable from that induced by the ligand. Here we provide evidence that in the COS-7 cell model EGFR transactivation via the muscarinic M2 receptor (M2R) is independent of MMPs and results in an incomplete EGFR signalling including ERK and Akt but not PLCgamma1. Using dominant-negative mutants of c-Src and Fyn and Src-deficient SYF cells as well as by co-immunoprecipitation studies, we can demonstrate that the M2R-mediated transactivation of EGFR specifically involves Fyn but not c-Src or Yes. This specific role of Fyn can be verified in SH-SY5Y human neuroblastoma cells with endogenously expressed M2 receptors.


Subject(s)
ErbB Receptors/genetics , Matrix Metalloproteinases/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , Receptor, Muscarinic M2/metabolism , Transcriptional Activation/genetics , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Mice , Phospholipase C gamma/metabolism , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-yes/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Signal Transduction
12.
Curr Pharm Des ; 10(16): 1937-58, 2004.
Article in English | MEDLINE | ID: mdl-15180530

ABSTRACT

In recent years, new strategies in cancer therapy have been developed targeting key signaling molecules in the receptor tyrosine kinase signal transduction pathway. In contrast, most therapeutical concepts to manipulate G protein-coupled receptors (GPCR)-mediated disorders are still limited to the use of receptor-specific agonists or antagonists. Visible progress in the understanding of GPCR signaling complexity, especially the detection of several families of highly target- and cell-specific regulator proteins of GPCRs, G proteins, and effector components may open new horizons to develop novel therapeutical concepts targeting GPCR signaling elements. Thus, this review will focus on different molecular levels that may be of particular interest in terms of new drug development such as: (i) GPCR subtypes, allosteric binding sites, dimerization and constitutive activity, the use of RAMPs (receptor-activity-modifying proteins) and RASSLs (receptor activated solely by synthetic ligands); (ii) AGS (activators of G protein signaling) and RGS (regulators of G protein signaling) proteins which modify G protein activity; (iii) the high diversity of isozymes involved in the generation, signal transmission, and degradation of second messenger molecules.


Subject(s)
Drug Delivery Systems/methods , Receptors, G-Protein-Coupled/agonists , Signal Transduction/drug effects , Signal Transduction/physiology , Animals , GTP-Binding Proteins/agonists , GTP-Binding Proteins/metabolism , Humans , Protein Binding/drug effects , Protein Binding/physiology , Receptors, G-Protein-Coupled/metabolism
13.
Br J Pharmacol ; 140(1): 13-22, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12967930

ABSTRACT

(1) Here, we introduce a beta-casomorphin-5-derived cyclic pentapeptide, cCD-2 (Tyr-cyclo[d-Orn-Tyr(Bzl)-Pro-Gly]), which inhibits the cell growth of a variety of human cancer cell lines. (2) This opioid-derived peptide possesses only low affinity for mu-receptors, but enhances the agonist binding to mu-receptors in vitro and potentiates the analgesic effect of morphin in vivo. The molecular mechanism of mu-receptor sensitization by cCD-2 is not yet known. (3) The antiproliferative effect of cCD-2 is independent of mu-, delta-, and kappa-receptors. (4) Using SH-SY5Y cells as model, we can demonstrate that cCD-2 specifically binds to somatostatin receptors and stimulates the activity of protein tyrosine phosphatases, which are early downstream targets of SST receptors. (5) In SH-SY5Y cells, cCD-2 specifically increases the activity of the cytosolic PTP SHP-2, stimulates the activity of mitogen-activated protein kinase (MAPK), and elevates the expression of the cyclin-dependent kinase inhibitor p21 (WAF1/Cip1), suggesting the involvement of SSTR1 receptor subtype in cCD-2 action in this cell type. (6) In COS-7 cells, for comparison, we found a stimulation of SHP-2 as well as SHP-1 in response to cCD-2. The activation of SHP-1, which is attributed to the SSTR2 receptor and negatively regulates the EGF receptor, corresponds with the ability of cCD-2 to inhibit the EGF-induced MAPK activation in COS-7 cells. (7) Our results show that in SH-SY5Y cells cCD-2 inhibits cell growth via the SSTR1 receptor-signalling pathway but may, in other cells, also use other SSTR subtypes and their signalling mechanisms. (8) cCD-2 represents a novel type of opioid-derived antiproliferative SST receptor agonist, which possesses low mu-receptor affinity but may induce mu-receptor sensitization and is structurally different from the hitherto known SST receptor agonists.


Subject(s)
Endorphins/pharmacology , Growth Inhibitors/pharmacology , Peptide Fragments/pharmacology , Receptors, Opioid, mu/metabolism , Receptors, Somatostatin/agonists , Cell Division/drug effects , Cell Division/physiology , Cell Line, Tumor , Dose-Response Relationship, Drug , Endorphins/chemistry , Endorphins/metabolism , Growth Inhibitors/chemistry , Growth Inhibitors/metabolism , Humans , Narcotic Antagonists/pharmacology , Pain Measurement/drug effects , Pain Measurement/methods , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Somatostatin/metabolism
14.
Int Immunopharmacol ; 2(13-14): 1763-70, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12489790

ABSTRACT

To study the intramolecular signal transduction, we performed single point and cassette mutations in transmembranal and intracellular regions of the bradykinin B2 receptor. We studied the influence of the two intramembranal Cys residues at positions 304 and 348, the role of Arg at position 177 in the highly conserved tripeptide sequence Asp-Arg-Tyr, the cytosolic G-protein binding area, and attempted to verify the general hypothesis of an ion tunnel-like interface in GPCRs. Wild type receptor, His-tagged receptor, and His-tagged mutant receptors were expressed in COS-7 cells and functionally compared by bradykinin-induced formation of inositolphosphate and arachidonic acid. To investigate the expression, all mutants were modified at the N-terminus by insertion of two successive His-tags and detected with an anti-poly-His antibody. Replacement of the second and third cytosolic loop by a loop from another membrane protein as well as single replacement of Arg at position 177 by Ala leads to a fully inactive receptor mutant without any ligand binding affinity and stimulatory activity. Mutants with replacement of Cys residues 304 and 348 by Ser showed only moderate effects. Regardless of the replacement of Asp 407 by Ala, the receptor is able to increase the agonist-induced levels of inositolphosphate and of arachidonic acid, indicating that our studies can not verify the postulated ion tunnel hypothesis.


Subject(s)
Receptors, Bradykinin/metabolism , Signal Transduction , Amino Acid Substitution , Animals , Arachidonic Acid/metabolism , Binding Sites , Blotting, Western , COS Cells , Chlorocebus aethiops , Mutagenesis, Site-Directed , Point Mutation , Radioligand Assay , Receptor, Bradykinin B2 , Receptors, Bradykinin/genetics , Transfection
15.
Peptides ; 23(8): 1503-6, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12182954

ABSTRACT

The cyclic somatostatin (SST) analogue, cyclo-(7-aminoheptanoyl-Phe-D-Trp-Lys-Thr[BZL]) (cSSTA), has been widely used as somatostatin antagonist. In the human neuroblastoma cell line SH-SY5Y the cyclopeptide acts as a somatostatin receptor agonist. Similar to SST, cSSTA inhibits cell proliferation, activates the protein tyrosine phosphatase SHP-2, and stimulates the activity of mitogen-activated protein kinase. These results suggest that in SH-SY5Y neuroblastoma cells somatostatin receptors may exist which exhibit altered antagonist binding properties.


Subject(s)
Receptors, Somatostatin/antagonists & inhibitors , Somatostatin/analogs & derivatives , Somatostatin/antagonists & inhibitors , Somatostatin/metabolism , DNA/biosynthesis , Humans , Mitogen-Activated Protein Kinases/drug effects , Neuroblastoma/drug therapy , Nucleic Acid Synthesis Inhibitors/metabolism , Nucleic Acid Synthesis Inhibitors/pharmacology , Protein Tyrosine Phosphatases/drug effects , Protein Tyrosine Phosphatases/metabolism , Somatostatin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...