Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Sci Rep ; 11(1): 19877, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34615939

ABSTRACT

ATP-dependent P2X3 receptors play a crucial role in the sensitization of nerve fibers and pathological pain pathways. They are also involved in pathways triggering cough and may contribute to the pathophysiology of endometriosis and overactive bladder. However, despite the strong therapeutic rationale for targeting P2X3 receptors, preliminary antagonists have been hampered by off-target effects, including severe taste disturbances associated with blocking the P2X2/3 receptor heterotrimer. Here we present a P2X3 receptor antagonist, eliapixant (BAY 1817080), which is both highly potent and selective for P2X3 over other P2X subtypes in vitro, including P2X2/3. We show that eliapixant reduces inflammatory pain in relevant animal models. We also provide the first in vivo experimental evidence that P2X3 antagonism reduces neurogenic inflammation, a phenomenon hypothesised to contribute to several diseases, including endometriosis. To test whether eliapixant could help treat endometriosis, we confirmed P2X3 expression on nerve fibers innervating human endometriotic lesions. We then demonstrate that eliapixant reduces vaginal hyperalgesia in an animal model of endometriosis-associated dyspareunia, even beyond treatment cessation. Our findings indicate that P2X3 antagonism could alleviate pain, including non-menstrual pelvic pain, and modify the underlying disease pathophysiology in women with endometriosis. Eliapixant is currently under clinical development for the treatment of disorders associated with hypersensitive nerve fibers.


Subject(s)
Nerve Fibers/drug effects , Nerve Fibers/metabolism , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X3/metabolism , Somatosensory Disorders/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Line , Disease Models, Animal , Female , Gene Expression , Humans , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Membrane Potentials/drug effects , Mice , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Rats , Receptors, Purinergic P2X3/genetics , Somatosensory Disorders/drug therapy , Somatosensory Disorders/etiology
2.
Sci Transl Med ; 13(608)2021 08 25.
Article in English | MEDLINE | ID: mdl-34433639

ABSTRACT

Endometriosis is a common chronic inflammatory condition causing pelvic pain and infertility in women, with limited treatment options and 50% heritability. We leveraged genetic analyses in two species with spontaneous endometriosis, humans and the rhesus macaque, to uncover treatment targets. We sequenced DNA from 32 human families contributing to a genetic linkage signal on chromosome 7p13-15 and observed significant overrepresentation of predicted deleterious low-frequency coding variants in NPSR1, the gene encoding neuropeptide S receptor 1, in cases (predominantly stage III/IV) versus controls (P = 7.8 × 10-4). Significant linkage to the region orthologous to human 7p13-15 was replicated in a pedigree of 849 rhesus macaques (P = 0.0095). Targeted association analyses in 3194 surgically confirmed, unrelated cases and 7060 controls revealed that a common insertion/deletion variant, rs142885915, was significantly associated with stage III/IV endometriosis (P = 5.2 × 10-5; odds ratio, 1.23; 95% CI, 1.09 to 1.39). Immunohistochemistry, qRT-PCR, and flow cytometry experiments demonstrated that NPSR1 was expressed in glandular epithelium from eutopic and ectopic endometrium, and on monocytes in peritoneal fluid. The NPSR1 inhibitor SHA 68R blocked NPSR1-mediated signaling, proinflammatory TNF-α release, and monocyte chemotaxis in vitro (P < 0.01), and led to a significant reduction of inflammatory cell infiltrate and abdominal pain (P < 0.05) in a mouse model of peritoneal inflammation as well as in a mouse model of endometriosis. We conclude that the NPSR1/NPS system is a genetically validated, nonhormonal target for the treatment of endometriosis with likely increased relevance to stage III/IV disease.


Subject(s)
Endometriosis , Receptors, G-Protein-Coupled/genetics , Animals , Endometriosis/drug therapy , Endometriosis/genetics , Endometrium , Female , Humans , Macaca mulatta , Mice , Tumor Necrosis Factor-alpha
4.
Sci Rep ; 10(1): 1495, 2020 01 30.
Article in English | MEDLINE | ID: mdl-32001775

ABSTRACT

Endometriosis is a common gynaecological disease of women in reproductive age, and is thought to arise from retrograde menstruation and implantation of endometrial tissue, mostly into the peritoneal cavity. The condition is characterized by a chronic, unresolved inflammatory process thereby contributing to pain as cardinal symptom in endometriosis. Elevated reactive oxygen species (ROS) and oxidative stress have been postulated as factors in endometriosis pathogenesis. We here set out for a systematic study to identify novel mechanisms and pathways relating to oxidative stress in ectopic peritoneal lesions. Using combined proteomic and transcriptomic approaches, we identified novel targets including upregulated pro-oxidative enzymes, such as amine oxidase 3/vascular adhesion protein 1 (AOC3/VAP1) as well as downregulated protective factors, in particular alkenal reductase PTGR1 and methionine sulfoxide reductase. Consistent with an altered ROS landscape, we observed hemoglobin / iron overload, ROS production and lipid peroxidation in ectopic lesions. ROS-derived 4-hydroxy-2-nonenal induced interleukin IL-8 release from monocytes. Notably, AOC3 inhibitors provoked analgesic effects in inflammatory pain models in vivo, suggesting potential translational applicability.


Subject(s)
Amine Oxidase (Copper-Containing)/metabolism , Cell Adhesion Molecules/metabolism , Endometriosis/metabolism , Peritoneal Diseases/metabolism , Aldehydes/metabolism , Allyl Compounds/pharmacology , Amine Oxidase (Copper-Containing)/antagonists & inhibitors , Analgesics/pharmacology , Animals , Biomarkers/metabolism , Cell Adhesion Molecules/antagonists & inhibitors , Disease Models, Animal , Endometriosis/genetics , Endometriosis/pathology , Female , Gene Expression Profiling , Heme/metabolism , Humans , Inflammation Mediators/metabolism , Interleukin-8/metabolism , Iron/metabolism , Lipid Peroxidation , Metabolic Networks and Pathways , Mice , Mice, Inbred BALB C , Myeloid Cells/pathology , Oxidative Stress , Peritoneal Diseases/genetics , Peritoneal Diseases/pathology , Phagocytosis , Sulfonamides/pharmacology
5.
Hum Reprod ; 28(8): 2253-64, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23739217

ABSTRACT

STUDY QUESTION: Does the novel progesterone receptor (PR) modulator BAY 1002670, based on its preclinical pharmacological profile, offer a potential novel treatment option for uterine fibroids? SUMMARY ANSWER: The newly synthesized BAY1002670 has proved to be a very potent, highly selective PR modulator in all in vitro and in vivo pharmacodynamics assays performed: it exhibits marked efficacy in an innovative humanized fibroid disease model, suggesting BAY 1002670 to be a very promising treatment option for uterine fibroids. WHAT IS KNOWN ALREADY: PR inhibiting ligands have shown clinical utility in a range of potential indications and applications. Despite the emergence of the first PR antagonist >30 years ago, no agent of this compound class has been authorized in any indication for long-term application. Among other reasons, suboptimal selectivity and safety profiles of previous candidates have led to discontinuation and modification of development programmes. STUDY, DESIGN, SIZE, DURATION: The preclinical studies include relevant in vitro and in vivo assays to clarify the properties of the PR modulator BAY 1002670 as well as a fibroid xenograft study to show directly the efficacy of BAY 1002670 on the human target tissue. PARTICIPANTS/MATERIAL, SETTING, METHODS: BAY 1002670 was tested for binding and transactivational activity towards different human steroid receptors. Activity of the compound in the corresponding in vivo models (rat, rabbit) was assessed. Furthermore, BAY 1002670 was tested in a disease model for uterine fibroids utilizing primary human tumour tissues as xenograft in immunodeficient mice treated with estradiol (E2) and progesterone (P). MAIN RESULT AND THE ROLE OF CHANCE: BAY1002670 in subnanomolar concentrations exhibits a highly selective binding profile and antagonistic activity for the PR. These properties are also reflected in its action in two progesterone-dependent animal models that assess the termination of pregnancy and endometrial transformation. Favourable selectivity towards other nuclear hormone receptors was demonstrated. No in vivo activity was found at the glucocorticoid, estrogenic and mineralocorticoid receptors with only weak anti-androgenic activity. In a human fibroid xenograft model BAY 1002670 showed a marked dose-dependent reduction of fibroid tumour weight gain of 95% at a dose of 3 mg/kg/day (P < 0.005). LIMITATIONS AND REASON FOR CAUTION: Selectivity and potency of BAY 1002670 have only been determined in vitro and in animal models so far. WIDER IMPLICATIONS OF THE FINDING: The PR modulator BAY 1002670 might offer a treatment option not only for uterine fibroids but also for other gynaecological indications. STUDY FUNDING/COMPETING INTEREST: The studies took place at Bayer Pharma AG. All authors are employees of Bayer Pharma AG. No external funding declared.


Subject(s)
Genital Diseases, Female/drug therapy , Receptors, Progesterone/drug effects , Steroids/pharmacology , Animals , Estradiol/pharmacology , Female , Heterografts/drug effects , Humans , Leiomyoma/drug therapy , Mice , Progesterone/pharmacology , Rabbits , Rats , Steroids/chemistry , Steroids/therapeutic use , Transcriptional Activation/drug effects
6.
Reprod Sci ; 20(1): 85-102, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22878529

ABSTRACT

We identified differentially expressed genes comparing peritoneal endometriosis lesions (n = 18), eutopic endometrium (n = 17), and peritoneum (n = 22) from the same patients with complete menstrual cycles using microarrays (54 675 probe sets) and immunohistochemistry. Peritoneal lesions and peritoneum demonstrated 3901 and 4973 significantly differentially expressed genes compared to eutopic endometrium, respectively. Peritoneal lesions significantly revealed no correlation with a specific menstrual cycle phase by gene expression and histopathology, exhibited low expressed proliferation genes, and constant levels of steroid hormone receptor genes. Tissue remodeling genes in cytoskeleton, smooth muscle contraction, cellular adhesion, tight junctions, and O-glycan biosynthesis were the most significant to lesions, including desmin and smooth muscle myosin heavy chain 11. Protein expression and location of desmin, alpha-actin, and h-caldesmon in peritoneal lesions discriminated between smooth muscle hyperplasia and metaplasia. Peritoneal lesions demonstrate no menstrual cycle phasing but constant steroid hormone receptor expression where a slow but steady growth is linked with tissue remodeling. Our study contributes to the molecular pathology of peritoneal endometriosis and will help to identify clinical targets for treatment and management.


Subject(s)
Endometriosis/genetics , Endometriosis/pathology , Endometrium/pathology , Menstrual Cycle/genetics , Peritoneum/pathology , Adult , Cohort Studies , Endometriosis/metabolism , Endometrium/metabolism , Female , Humans , Menstrual Cycle/metabolism , Middle Aged , Peritoneum/metabolism , Principal Component Analysis , Young Adult
7.
Methods Mol Biol ; 327: 85-97, 2006.
Article in English | MEDLINE | ID: mdl-16780214

ABSTRACT

Interreceptor cross-talk has emerged as a general concept in cellular signaling cascades. Therein epidermal growth factor receptor (EGFR) signal transactivation represents the so far best investigated cross-talk mechanism comprising heterogeneous receptor families. In this signaling process G protein-coupled receptor (GPCR) stimulation induces phosphorylation of the EGFR, combining the broad diversity of GPCRs with the potent signaling capacities of this receptor tyrosine kinase. Early reports attributed this transactivation mechanism to solely intracellular pathways as no EGF-like ligands could be detected in conditioned media of GPCR agonist-stimulated cells. However, Prenzel and colleagues demonstrated the involvement of metalloproteinase-mediated release of EGF-like ligands as the predominant mechanism of EGFR signal transactivation, providing a point of convergence for different intracellular effector proteins. Since this discovery, numerous investigations revealed the broad relevance of metalloproteinase-mediated ligand-dependent EGFR signal transactivation for coupling GPCRs to various cellular signaling responses. Here we describe methods to investigate GPCR-stimulated EGFR signal transactivation allowing the identification of both the EGF-like ligands and the metalloproteinases involved.


Subject(s)
ErbB Receptors/metabolism , Signal Transduction , ADAM Proteins/metabolism , ADAM17 Protein , Animals , COS Cells , Chlorocebus aethiops , Culture Media , Epidermal Growth Factor/metabolism , Flow Cytometry , Humans , Ligands , Oligonucleotides/metabolism , RNA, Small Interfering/genetics
8.
Biol Chem ; 386(9): 845-55, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16164409

ABSTRACT

The epidermal growth factor receptor (EGFR) plays a key role in the regulation of important cellular processes under normal and pathophysiological conditions such as cancer. In human mammary carcinomas the EGFR is involved in regulating cell growth, survival, migration and metastasis and its activation correlates with the lack of response in hormone therapy. Here, we demonstrate in oestrogen receptor-positive and -negative human breast cancer cells and primary mammary epithelial cells a cross-communication between G protein-coupled receptors (GPCRs) and the EGFR. We present evidence that specific inhibition of ADAM15 or TACE blocks GPCR-induced and proHB-EGF-mediated EGFR tyrosine phosphorylation, downstream mitogenic signalling and cell migration. Notably, activation of the PI3K downstream mediator PKB/Akt by GPCR ligands involves the activity of sphingosine kinase (SPHK) and is independent of EGFR signal transactivation. We conclude that GPCR-induced chemotaxis of breast cancer cells is mediated by EGFR-dependent and -independent signalling pathways, with both parallel pathways having to act in concert to achieve a complete migratory response.


Subject(s)
Breast Neoplasms/pathology , Cell Movement/physiology , ErbB Receptors/physiology , Receptors, G-Protein-Coupled/physiology , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/genetics , ADAM Proteins/metabolism , ADAM17 Protein , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Enzyme Activation , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Metalloproteases/antagonists & inhibitors , Metalloproteases/metabolism , Mice , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Phospholipids/pharmacology , Phosphorylation , Pregnancy , Proto-Oncogene Proteins c-akt/metabolism , Receptor Cross-Talk/physiology , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Shc Signaling Adaptor Proteins , Signal Transduction/physiology , Src Homology 2 Domain-Containing, Transforming Protein 1 , Transfection , Tumor Cells, Cultured
9.
Mol Cell Biol ; 24(12): 5172-83, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15169883

ABSTRACT

Mammalian cells respond to environmental stress by activating a variety of protein kinases critical for cellular signal transmission, such as the epidermal growth factor receptor (EGFR) tyrosine kinase and different members of the mitogen-activated protein kinase (MAPK) family. EGFR activation by stress stimuli was previously thought to occur independently of stimulation by extracellular ligands. Here, we provide evidence that osmotic and oxidative stresses induce a metalloprotease activity leading to cell surface cleavage of pro-heparin-binding EGF (pro-HB-EGF) and subsequent EGFR activation. This ligand-dependent EGFR signal resulted from stress-induced activation of the MAPK p38 in human carcinoma cells and was mediated by the metalloproteases ADAM9, -10, and -17. Furthermore, stress-induced EGFR activation induced downstream signaling through the MAPKs extracellular signal-regulated kinases 1 and 2 and JNK. Interestingly, apoptosis induced by treatment of tumor cells with doxorubicin was strongly enhanced by blocking HB-EGF function. Together, our data provide novel insights into the mammalian stress response, suggesting a broad mechanistic relevance of a p38-ADAM-HB-EGF-EGFR-dependent pathway and its potential significance for tumor cells in evasion of chemotherapeutic agent-induced apoptosis.


Subject(s)
Epidermal Growth Factor/metabolism , Heparin/metabolism , Metalloendopeptidases/metabolism , Animals , Apoptosis/drug effects , Base Sequence , COS Cells , Cell Line, Tumor , Doxorubicin/pharmacology , Gene Silencing , Heparin-binding EGF-like Growth Factor , Humans , Intercellular Signaling Peptides and Proteins , Kinetics , MAP Kinase Signaling System , Metalloendopeptidases/genetics , Osmotic Pressure , Oxidative Stress , Phosphorylation , RNA, Neoplasm/genetics , RNA, Small Interfering/genetics , Signal Transduction
10.
J Biol Chem ; 279(28): 28970-8, 2004 Jul 09.
Article in English | MEDLINE | ID: mdl-15123705

ABSTRACT

Cross-communication between the Met receptor tyrosine kinase and the epidermal growth factor receptor (EGFR) has been proposed to involve direct association of both receptors and EGFR kinase-dependent phosphorylation. Here, we demonstrate that in human hepatocellular and pancreatic carcinoma cells the Met receptor becomes tyrosine phosphorylated not only upon EGF stimulation but also in response to G protein-coupled receptor (GPCR) agonists. Whereas specific inhibition of the EGFR kinase activity blocked EGF- but not GPCR agonist-induced Met receptor transactivation, it was abrogated in the presence of a reducing agent or treatment of cells with a NADPH oxidase inhibitor. Both GPCR ligands and EGF are further shown to increase the level of reactive oxygen species within the cell. Interestingly, stimulation of the Met receptor by either GPCR agonists, EGF or its cognate ligand HGF, resulted in release of Met-associated beta-catenin and in its Met-dependent translocation into the nucleus, as analyzed by small interfering RNA-mediated knockdown of the Met receptor. Our results provide a new molecular explanation for cell surface receptor cross-talk involving the Met receptor and thereby link the wide diversity of GPCRs and the EGFR to the oncogenic potential of Met signaling in human carcinoma cells.


Subject(s)
ErbB Receptors/metabolism , Proteins/metabolism , Proto-Oncogene Proteins , Reactive Oxygen Species/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Growth Factor , Transcriptional Activation , Animals , Carcinoma/metabolism , Cell Fractionation , Cell Line, Tumor , Cytoskeletal Proteins/metabolism , Enzyme Inhibitors/metabolism , Epidermal Growth Factor/metabolism , Humans , Ligands , MAP Kinase Signaling System/physiology , NADPH Oxidases/metabolism , Phosphorylation , Proteins/genetics , Proto-Oncogene Proteins c-met , RNA, Small Interfering/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Trans-Activators/metabolism , beta Catenin
12.
Cancer Res ; 64(6): 1943-50, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-15026328

ABSTRACT

Cannabinoids, the active components of marijuana and their endogenous counterparts were reported as useful analgetic agents to accompany primary cancer treatment by preventing nausea, vomiting, and pain and by stimulating appetite. Moreover, they have been shown to inhibit cell growth and to induce apoptosis in tumor cells. Here, we demonstrate that anandamide, Delta(9)-tetrahydrocannabinol (THC), HU-210, and Win55,212-2 promote mitogenic kinase signaling in cancer cells. Treatment of the glioblastoma cell line U373-MG and the lung carcinoma cell line NCI-H292 with nanomolar concentrations of THC led to accelerated cell proliferation that was completely dependent on metalloprotease and epidermal growth factor receptor (EGFR) activity. EGFR signal transactivation was identified as the mechanistic link between cannabinoid receptors and the activation of the mitogen-activated protein kinases extracellular signal-regulated kinase 1/2 as well as prosurvival protein kinase B (Akt/PKB) signaling. Depending on the cellular context, signal cross-communication was mediated by shedding of proAmphiregulin (proAR) and/or proHeparin-binding epidermal growth factor-like growth factor (proHB-EGF) by tumor necrosis factor alpha converting enzyme (TACE/ADAM17). Taken together, our data show that concentrations of THC comparable with those detected in the serum of patients after THC administration accelerate proliferation of cancer cells instead of apoptosis and thereby contribute to cancer progression in patients.


Subject(s)
Apoptosis/drug effects , Cannabinoids/pharmacology , Dronabinol/analogs & derivatives , ErbB Receptors/metabolism , Metalloendopeptidases/metabolism , Transcriptional Activation , ADAM Proteins , ADAM17 Protein , Amphiregulin , Benzoxazines , Cell Division/drug effects , Dronabinol/pharmacology , EGF Family of Proteins , Enzyme Activation/drug effects , Epidermal Growth Factor/metabolism , ErbB Receptors/genetics , Glioblastoma/enzymology , Glioblastoma/pathology , Glycoproteins/metabolism , Heparin-binding EGF-like Growth Factor , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Morpholines/pharmacology , Naphthalenes/pharmacology , Phosphorylation , Proto-Oncogene Proteins/metabolism , RNA Interference , Signal Transduction , Tumor Cells, Cultured
13.
Discov Med ; 4(22): 166-71, 2004 Jun.
Article in English | MEDLINE | ID: mdl-20704980

ABSTRACT

Extract: Molecular communication is essential for the coordinated development and life of multicellular organisms. This process involves sending, receiving and promoting signals by means of elaborate signal transduction networks. Important players for these processes are cell surface receptors which transmit signals across the cell's outer barrier, the cell membrane. The so-called receptor tyrosine kinases (RTKs) play a central role within this group of proteins by controlling a wide array of biological functions such as cell migration, proliferation, survival or differentiation. Concomitantly, deregulated signal transduction leads to aberrant cellular behavior and, as a consequence, can induce or contribute to various pathophysiological disorders such as cancer. Based on this rationale RTKs have been discovered as prime targets for therapeutic intervention and the first drugs that have been approved demonstrate the potential of this approach. A kinase is an enzyme that catalyzes the addition of a phosphoryl group to a molecule, an event called phosphorylation. Tyrosine kinase adds a phosphoryl group to the tyrosine residue of a protein.

14.
Curr Opin Chem Biol ; 7(4): 490-5, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12941424

ABSTRACT

Identification of the key role of protein kinases as potential oncoproteins has led to the emergence of a new era of target-directed therapies. Among a variety of novel therapeutic strategies two have shown the most promise and led to a variety of therapeutic agents in clinical development. One approach utilises humanised monoclonal antibodies generated against the extracellular domain of transmembrane protein kinases. The second approach is the generation of small molecule ATP analogues targeting the kinase domain itself. The approval of agents such as Herceptin for the treatment of advanced breast cancer and Gleevec for chronic myelogenous leukemia and gastrointestinal stromal tumours are the first examples of gene-based cancer drugs and represent the first example of a novel strategy in anti-cancer therapy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Piperazines/therapeutic use , Pyrimidines/therapeutic use , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacology , Benzamides , Cell Cycle/drug effects , Drug Delivery Systems , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Humans , Imatinib Mesylate , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/metabolism , Trastuzumab
15.
EMBO J ; 22(10): 2411-21, 2003 May 15.
Article in English | MEDLINE | ID: mdl-12743035

ABSTRACT

Communication between G protein-coupled receptor (GPCR) and epidermal growth factor receptor (EGFR) signalling systems involves cell surface proteolysis of EGF-like precursors. The underlying mechanisms of EGFR signal transactivation pathways, however, are largely unknown. We demonstrate that in squamous cell carcinoma cells, stimulation with the GPCR agonists LPA or carbachol specifically results in metalloprotease cleavage and release of amphiregulin (AR). Moreover, AR gene silencing by siRNA or inhibition of AR biological activity by neutralizing antibodies and heparin prevents GPCR-induced EGFR tyrosine phosphorylation, downstream mitogenic signalling events, cell proliferation, migration and activation of the survival mediator Akt/PKB. Therefore, despite some functional redundancy among EGF family ligands, the present study reveals a distinct and essential role for AR in GPCR-triggered cellular responses. Furthermore, we present evidence that blockade of the metalloprotease-disintegrin tumour necrosis factor-alpha-converting enzyme (TACE) by the tissue inhibitor of metalloprotease-3, a dominant-negative TACE mutant or RNA interference suppresses GPCR-stimulated AR release, EGFR activation and downstream events. Thus, TACE can function as an effector of GPCR-mediated signalling and represents a key element of the cellular receptor cross-talk network.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Cell Movement/physiology , GTP-Binding Proteins/metabolism , Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Metalloendopeptidases/metabolism , Protein Precursors/metabolism , Protein Serine-Threonine Kinases , Receptors, Cell Surface/metabolism , ADAM Proteins , ADAM17 Protein , Amphiregulin , Antibodies/metabolism , Carbachol/metabolism , Carcinoma, Squamous Cell/pathology , Cell Division/physiology , EGF Family of Proteins , Enzyme Activation , ErbB Receptors/metabolism , Humans , Ligands , Lysophospholipids/metabolism , Membrane Proteins/metabolism , Metalloendopeptidases/genetics , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , RNA, Small Interfering/metabolism , Signal Transduction/physiology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...