Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Neurourol Urodyn ; 38(5): 1250-1260, 2019 06.
Article in English | MEDLINE | ID: mdl-30989724

ABSTRACT

AIMS: Central sensitization playsimportant roles in cyclophosphamide (CYP)-induced cystitis. In addition, as a visceral pain, CYP-induced chronic pain shares common pathophysiological mechanisms with neuropathic pain. Previous studies demonstrated that neuregulin-1 (Nrg1)-ErbB signaling contributes to neuropathic pain, but whether and how this signaling influences mechanical allodynia in CYP-induced cystitis is unclear. This study aimed to determine whether and how Nrg1-ErbB signaling modulates mechanical allodynia in a CYP-induced cystitis rat model. METHODS: Systemic injection with CYP was used to establish a rat model of bladder pain syndrome/interstitial cystitis (BPS/IC). An irreversible ErbB family receptor inhibitor, PD168393, and exogenous Nrg1 were intrathecally injected to modulate Nrg1-ErbB signaling. Mechanical allodynia in the lower abdomen was assessed with von-Frey filaments using the up-down method. Western blot analysis and immunofluorescence staining were used to measure the expression of Nrg1-ErbB signaling, Iba-1, p-p38, and IL-1ß in the L6-S1 spinal dorsal horn (SDH). RESULTS: We observed upregulation of Nrg1-ErbB signaling as well as overexpression of the microglia activation markers Iba-1 and p-p38 and the proinflammatory factor, interleukin-1ß (IL-1ß), in the SDH of the cystitis group. Further, treatment with PD168393 attenuated mechanical allodynia in CYP-induced cystitis and inhibited microglia activation, leading to decreased production of IL-1ß. The inhibitor PD168393 reversed the algesic effect of exogenous Nrg1 on the cystitis model. CONCLUSIONS: Nrg1-ErbB signaling may promote microglia activation, contributing to mechanical allodynia of CYP-induced cystitis. Our study showed that modulation of Nrg1-ErbB signaling may have therapeutic value for treating pain symptoms in BPS/IC.


Subject(s)
Cystitis/chemically induced , Hyperalgesia/chemically induced , Microglia , Neuregulin-1/physiology , Oncogene Proteins v-erbB/physiology , Animals , Cystitis/complications , Cystitis/drug therapy , Female , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Injections, Spinal , Macrophage Activation , Quinazolines/pharmacology , Quinazolines/therapeutic use , Rats , Rats, Sprague-Dawley , Signal Transduction
2.
Int J Clin Exp Pathol ; 7(4): 1449-58, 2014.
Article in English | MEDLINE | ID: mdl-24817940

ABSTRACT

Paclitaxel (PTX) has shown encouraging activity in the treatment of advanced gastric cancer (GC). However, the fact that more than half of GC patients respond poorly to PTX-based chemotherapies demonstrates the urgent need for biomarkers of PTX sensitivity in GC patients. In the present work, three GC cell lines (BGC-823, HGC-27 and NCI-N87) with different sensitivities to PTX were subjected to DNA microarray analysis. The significantly differentially expressed genes and microRNAs (miRs) were identified and pathway signatures for PTX sensitivity were proposed. Ingenuity Pathway Analysis results showed that the differentially expressed genes were mainly enriched in the ErbB signaling pathway and other pathways. Additionally, the AKT/ERK signaling pathway, which is the pathway downstream of ErbB, was predicted to be active in PTX-resistant GC cell lines. ErbB3 overexpression and AKT/ERK activation in PTX-resistant cell lines were validated, respectively, by quantitative PCR and immunoblotting. Furthermore, 10 miRs were dramatically differently expressed in the three GC cell lines, and a miR-gene network was constructed from these data. Our work uncovered a reliable signature for PTX sensitivity in GC and potential therapeutic targets for GC treatments.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents, Phytogenic/therapeutic use , Drug Resistance, Neoplasm/physiology , Extracellular Signal-Regulated MAP Kinases/physiology , Paclitaxel/therapeutic use , Proto-Oncogene Proteins c-akt/physiology , Stomach Neoplasms/drug therapy , Adenocarcinoma/pathology , Adenocarcinoma/physiopathology , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/genetics , Gene Expression Regulation, Neoplastic/physiology , Humans , Oligonucleotide Array Sequence Analysis , Oncogene Proteins v-erbB/genetics , Oncogene Proteins v-erbB/physiology , Paclitaxel/pharmacology , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Stomach Neoplasms/pathology , Stomach Neoplasms/physiopathology , Treatment Failure
3.
Cold Spring Harb Perspect Biol ; 6(4): a008961, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24691959

ABSTRACT

We review the states of the ErbB family of receptor tyrosine kinases (RTKs), primarily the EGF receptor (EGFR, ErbB1, HER1) and the orphan receptor ErbB2 as they exist in living mammalian cells, focusing on four main aspects: (1) aggregation state and distribution in the plasma membrane; (2) conformational features of the receptors situated in the plasma membrane, compared to the crystallographic structures of the isolated extracellular domains; (3) coupling of receptor disposition on filopodia with the transduction of signaling ligand gradients; and (4) ligand-independent receptor activation by application of a magnetic field.


Subject(s)
Cell Membrane/metabolism , Oncogene Proteins v-erbB/physiology , Animals , Crystallography, X-Ray , Mammals/metabolism , Oncogene Proteins v-erbB/chemistry , Oncogene Proteins v-erbB/metabolism , Protein Aggregates , Pseudopodia/metabolism , Signal Transduction
4.
Mol Pharmacol ; 83(4): 882-93, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23371912

ABSTRACT

Our laboratory has previously shown that some gefitinib-insensitive head and neck squamous cell carcinoma (HNSCC) cell lines exhibit dominant autocrine fibroblast growth factor receptor (FGFR) signaling. Herein, we deployed a whole-genome loss-of-function screen to identify genes whose knockdown potentiated the inhibitory effect of the FGFR inhibitor, AZ8010, in HNSCC cell lines. Three HNSCC cell lines expressing a genome-wide small hairpin RNA (shRNA) library were treated with AZ8010 and the abundance of shRNA sequences was assessed by deep sequencing. Under-represented shRNAs in treated cells are expected to target genes important for survival with AZ8010 treatment. Synthetic lethal hits were validated with specific inhibitors and independent shRNAs. We found that multiple alternate receptors provided protection from FGFR inhibition, including receptor tyrosine kinases (RTKs), v-erb-b2 erythroblastic leukemia viral oncogene homolog 2 (ERBB2), and hepatocyte growth factor receptor (MET). We showed that specific knockdown of either ERBB2 or MET in combination with FGFR inhibition led to increased inhibition of growth relative to FGFR tyrosine kinase inhibitor (TKI) treatment alone. These results were confirmed using specific small molecule inhibitors of either ERBB family members or MET. Moreover, the triple combination of FGFR, MET, and ERBB family inhibitors showed the largest inhibition of growth and induction of apoptosis compared with the double combinations. These results reveal a role for alternate RTKs in maintaining progrowth and survival signaling in HNSCC cells in the setting of FGFR inhibition. Thus, improved therapies for HNSCC patients could involve rationally designed combinations of TKIs targeting FGFR, ERBB family members, and MET.


Subject(s)
ErbB Receptors/physiology , Head and Neck Neoplasms/pathology , Oncogene Proteins v-erbB/physiology , Proto-Oncogene Proteins c-met/physiology , Receptor, ErbB-2/physiology , Receptors, Fibroblast Growth Factor/physiology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/physiology , ErbB Receptors/antagonists & inhibitors , Gene Regulatory Networks/drug effects , Gene Regulatory Networks/physiology , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Humans , Oncogene Proteins v-erbB/antagonists & inhibitors , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Quinazolines/pharmacology , Quinazolines/therapeutic use , Receptor Protein-Tyrosine Kinases/physiology , Receptor, ErbB-2/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/antagonists & inhibitors
5.
Development ; 140(5): 1003-13, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23364329

ABSTRACT

Pigment cells in vertebrates are derived from the neural crest (NC), a pluripotent and migratory embryonic cell population. In fishes, larval melanophores develop during embryogenesis directly from NC cells migrating along dorsolateral and ventromedial paths. The embryonic origin of the melanophores that emerge during juvenile development in the skin to contribute to the striking colour patterns of adult fishes remains elusive. We have identified a small set of melanophore progenitor cells (MPs) in the zebrafish (Danio rerio, Cyprinidae) that is established within the first 2 days of embryonic development in close association with the segmentally reiterated dorsal root ganglia (DRGs). Lineage analysis and 4D in vivo imaging indicate that progeny of these embryonic MPs spread segmentally, giving rise to the melanophores that create the adult melanophore stripes. Upon depletion of larval melanophores by morpholino knockdown of Mitfa, the embryonic MPs are prematurely activated; their progeny migrate along the spinal nerves restoring the larval pattern and giving rise to postembryonic MPs associated with the spinal nerves. Mutational or chemical inhibition of ErbB receptors blocks all early NC migration along the ventromedial path, causing a loss of DRGs and embryonic MPs. We show that the sparse like (slk) mutant lacks larval and metamorphic melanophores and identify kit ligand a (kitlga) as the underlying gene. Our data suggest that kitlga is required for the establishment or survival of embryonic MPs. We propose a model in which DRGs provide a niche for the stem cells of adult melanophores.


Subject(s)
Cell Lineage/genetics , Embryonic Stem Cells/physiology , Melanophores/physiology , Oncogene Proteins v-erbB/physiology , Proto-Oncogene Proteins c-kit/physiology , Zebrafish/embryology , Age Factors , Animals , Animals, Genetically Modified , Cell Movement/genetics , Cell Movement/physiology , Embryo, Nonmammalian , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental/drug effects , Melanophores/metabolism , Morpholinos/pharmacology , Motor Neurons/metabolism , Motor Neurons/physiology , Oncogene Proteins v-erbB/genetics , Oncogene Proteins v-erbB/metabolism , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish/physiology
6.
Circulation ; 125(24): 2993-3003, 2012 Jun 19.
Article in English | MEDLINE | ID: mdl-22610965

ABSTRACT

BACKGROUND: Chronic ß-adrenergic receptor (ß-AR) overstimulation, a hallmark of heart failure, is associated with increased cardiac expression of matrix metalloproteinases (MMPs). MMP-1 has been shown to cleave and activate the protease-activated receptor 1 (PAR1) in noncardiac cells. In the present study, we hypothesized that ß-AR stimulation would result in MMP-dependent PAR1 transactivation in cardiac cells. METHODS AND RESULTS: ß-AR stimulation of neonatal rat ventricular myocytes (NRVMs) or cardiac fibroblasts with isoproterenol transduced with an alkaline phosphatase-tagged PAR1 elicited a significant increase in alkaline phosphatase-PAR1 cleavage. This isoproterenol-dependent cleavage was significantly reduced by the broad-spectrum MMP inhibitor GM6001. Importantly, specific MMP-13 inhibitors also decreased alkaline phosphatase-PAR1 cleavage in isoproterenol-stimulated NRVMs, as well as in NRVMs stimulated with conditioned medium from isoproterenol-stimulated cardiac fibroblasts. Moreover, we found that recombinant MMP-13 stimulation cleaved alkaline phosphatase-PAR1 in NRVMs at DPRS(42)↓(43)FLLRN. This also led to the activation of the ERK1/2 pathway through Gαq in NRVMs and via the Gαq/ErbB receptor pathways in cardiac fibroblasts. MMP-13 elicited similar levels of ERK1/2 activation but lower levels of generation of inositol phosphates in comparison to thrombin. Finally, we demonstrated that either PAR1 genetic ablation or pharmacological inhibition of MMP-13 prevented isoproterenol-dependent cardiac dysfunction in mice. CONCLUSIONS: In this study, we demonstrate that ß-AR stimulation leads to MMP-13 transactivation of PAR1 in both cardiac fibroblasts and cardiomyocytes and that this likely contributes to pathological activation of Gαq and ErbB receptor-dependent pathways in the heart. We propose that this mechanism may underlie the development of ß-AR overstimulation-dependent cardiac dysfunction.


Subject(s)
Matrix Metalloproteinase 13/physiology , Myocytes, Cardiac/metabolism , Receptor, PAR-1/metabolism , Receptors, Adrenergic, beta/physiology , Transcriptional Activation , Animals , Extracellular Signal-Regulated MAP Kinases/physiology , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Humans , Male , Mice , Middle Aged , Oncogene Proteins v-erbB/physiology , Signal Transduction
7.
Biochem Soc Trans ; 39(2): 568-73, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21428941

ABSTRACT

Integrins and growth factor receptors of the ErbB family are involved in the regulation of cellular interactions with the extracellular microenvironment. Cross-talk between these two groups of transmembrane receptors is essential for cellular responses and can be regulated through the formation of multimolecular complexes. Tetraspanins as facilitators and building blocks of specialized microdomains may be involved in this process. In the present study, we demonstrated that, in contrast with previous reports, integrin-mediated adhesion did not stimulate ligand-independent activation of ErbB receptors in epithelial cells. However, integrin-dependent adhesion potentiated ligand-induced activation of EGFR (epidermal growth factor receptor) and ErbB2 and facilitated receptor homo- and hetero-dimerization. The actin cytoskeleton appeared to play a critical role in this phenomenon.


Subject(s)
Extracellular Matrix/metabolism , Oncogene Proteins v-erbB/physiology , Animals , Cell Adhesion/physiology , ErbB Receptors/genetics , ErbB Receptors/metabolism , ErbB Receptors/physiology , Extracellular Matrix/physiology , Humans , Integrins/metabolism , Integrins/physiology , Multigene Family/physiology , Oncogene Proteins v-erbB/genetics , Oncogene Proteins v-erbB/metabolism , Receptor Cross-Talk/physiology , Receptors, Growth Factor/genetics , Receptors, Growth Factor/metabolism , Receptors, Growth Factor/physiology
8.
Acta Med Port ; 24 Suppl 4: 1009-20, 2011 Dec.
Article in Portuguese | MEDLINE | ID: mdl-22863512

ABSTRACT

The family of Neuregulins (NRG), growth factors like epidermal growth factor, is known to induce growth and differentiation of epithelial, glial, neuronal, and skeletal muscle cells. This family comprises four members, being NRG1 the most largely studied, particularly at the cardiovascular level. The biological effects of NRG1 in the adult heart are mediated by the tyrosine kinase receptors ErbB. In the adult heart, NRG1 is expressed by cells of the endocardial endothelium and the cardiac microvascular endothelium, and the receptors ErbB2/ErbB4 are expressed by ventricular cardiomyocytes and are located in T-tubule system and intercalated disks in close proximity to the system components of excitation-contraction coupling. The importance of the NRG/ErbB signaling axis at the cardiovascular level became evident after discovering that patients treated with trastuzumab (inhibitory antibody against ErbB2, used in the treatment of breast cancer) can develop ventricular dysfunction and have higher risk of cardiomyopathy when co-administered with anthracyclines. Subsequent studies in vitro and in vivo have clarified the effects and the respective signaling pathways associated with the NRG/ErbB system in the adult heart. Some cardiovascular functions of the NRG1/ErbB system have been described at the vascular (stimulation of angiogenesis and ateroprotector effect) and myocardium level (negative inotropic effect) as well as effect on the survival, cell growth and organization of the cardiomyocytes (myofibrillar organization and cell-to-cell contact between cardiomyocytes). Furthermore, the interaction of this system with other neurohumoral mediators has been studied. Thus, there seems to be a physiological role in modulating the sympathovagal balance and an interaction with endothelin-1 signaling. All these effects result from the activation of different intracellular signaling cascades, as a consequence of the binding of NRG1 to ErbB receptors. Some cardiac signaling pathways identified until now include molecules such as MEK / Erk 1/2, phosphatidylinositol 3-kinase/ Akt, focal adhesion kinase, Gab (Grb-2-associated binder) family, vascular endothelial growth factor and NO production by endothelial nitric oxide synthase. Thus, the aim of this paper was to make an up-to-date review of existing information on NRG1/ErbB signaling axis, with particular focus on its cardiovascular effects.


Subject(s)
Heart/physiology , Neuregulin-1/physiology , Oncogene Proteins v-erbB/physiology , Humans
10.
Proc Natl Acad Sci U S A ; 106(11): 4507-12, 2009 Mar 17.
Article in English | MEDLINE | ID: mdl-19240213

ABSTRACT

Neuregulin-1 (NRG1) and its ErbB2/B4 receptors are encoded by candidate susceptibility genes for schizophrenia, yet the essential functions of NRG1 signaling in the CNS are still unclear. Using CRE/LOX technology, we have inactivated ErbB2/B4-mediated NRG1 signaling specifically in the CNS. In contrast to expectations, cell layers in the cerebral cortex, hippocampus, and cerebellum develop normally in the mutant mice. Instead, loss of ErbB2/B4 impairs dendritic spine maturation and perturbs interactions of postsynaptic scaffold proteins with glutamate receptors. Conversely, increased NRG1 levels promote spine maturation. ErbB2/B4-deficient mice show increased aggression and reduced prepulse inhibition. Treatment with the antipsychotic drug clozapine reverses the behavioral and spine defects. We conclude that ErbB2/B4-mediated NRG1 signaling modulates dendritic spine maturation, and that defects at glutamatergic synapses likely contribute to the behavioral abnormalities in ErbB2/B4-deficient mice.


Subject(s)
Cerebral Cortex/cytology , Dendritic Spines/pathology , Nerve Tissue Proteins/physiology , Receptor, ErbB-2/physiology , Signal Transduction , Adaptor Proteins, Signal Transducing , Animals , Antipsychotic Agents/pharmacology , Central Nervous System , Clozapine/pharmacology , Mice , Mice, Knockout , Nerve Tissue Proteins/deficiency , Neuregulin-1 , Oncogene Proteins v-erbB/deficiency , Oncogene Proteins v-erbB/physiology , Receptors, Glutamate
11.
J Neurosci ; 28(10): 2624-32, 2008 Mar 05.
Article in English | MEDLINE | ID: mdl-18322105

ABSTRACT

Cyclin-dependent kinase 5 (cdk5) and glycogen synthase kinase 3beta (GSK3beta) have been implicated in pathogenic processes associated with Alzheimer's disease because both kinases regulate tau hyperphosphorylation and enhance amyloid precursor protein (APP) processing leading to an increase in amyloid beta (Abeta) production. Here we show that young p25 overexpressing mice have enhanced cdk5 activity but reduced GSK3beta activity attributable to phosphorylation at the inhibitory GSK3beta-serine 9 (GSK3beta-S9) site. Phosphorylation at this site was mediated by enhanced activity of the neuregulin receptor complex, ErbB, and activation of the downstream phosphatidylinositol 3 kinase/Akt pathway. Young p25 mice had elevated Abeta peptide levels, but phospho-tau levels were decreased overall. Thus, cdk5 appears to play a dominant role in the regulation of amyloidogenic APP processing, whereas GSK3beta plays a dominant role in overall tau phosphorylation. In older mice, GSK3beta inhibitory phosphorylation at S9 was reduced relative to young mice. Abeta peptide levels were still elevated but phospho-tau levels were either unchanged or showed a trend to increase, suggesting that GSK3beta activity increases with aging. Inhibition of cdk5 by a specific inhibitor reduced cdk5 activity in p25 mice, leading to reduced Abeta production in both young and old mice. However, in young mice, cdk5 inhibition reversed GSK3beta inhibition, leading to an increase in overall tau phosphorylation. When cdk5 inhibitor was administered to very old, nontransgenic mice, inhibition of cdk5 reduced Abeta levels, and phospho-tau levels showed a trend to increase. Thus, cdk5 inhibitors may not be effective in targeting tau phosphorylation in the elderly.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Cyclin-Dependent Kinase 5/metabolism , Glycogen Synthase Kinase 3/metabolism , Neuregulins/physiology , Protein Interaction Mapping , Signal Transduction/physiology , tau Proteins/metabolism , Age Factors , Amyloid beta-Protein Precursor/genetics , Animals , Cell Line, Tumor , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Cyclin-Dependent Kinase 5/genetics , Down-Regulation/physiology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Humans , Mice , Mice, Transgenic , Nerve Tissue Proteins/physiology , Neuregulins/genetics , Neuregulins/metabolism , Oncogene Proteins v-erbB/physiology , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation , Phosphotransferases , Protein Kinase Inhibitors/pharmacology , tau Proteins/genetics
12.
Cell Mol Life Sci ; 64(24): 3159-84, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17955178

ABSTRACT

During its lifetime, the mammary gland undergoes many phases of development and differentiation. Much of this occurs during puberty, when the ductal epithelium expands by branching morphogenesis, invading the surrounding fat pad to form an organised mammary tree. Throughout its existence, the epithelium will go through several cycles of proliferation and cell death during pregnancy, lactation and involution. Many of the signalling mechanisms which control the initial invasion of the fat pad by the epithelium, and regulate its continuing plasticity, can be harnessed or corrupted by tumour cells in order to support their aberrant growth and progression towards invasion. This is true not just for the epithelial cells themselves but also for cells in the surrounding microenvironment, including fibroblasts, macrophages and adipocytes. This review examines the complex web of signalling and adhesion interactions controlling branching morphogenesis, and how their alteration can promote malignancy. Current in vivo and in vitro mammary gland models are also discussed. (Part of a Multi-author Review).


Subject(s)
Breast Neoplasms/genetics , Mammary Glands, Human/growth & development , Animals , Breast Neoplasms/metabolism , Cell Adhesion/physiology , Epidermal Growth Factor/genetics , Epidermal Growth Factor/physiology , Humans , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Mammary Glands, Human/metabolism , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Matrix Metalloproteinases/physiology , Models, Biological , Neoplasm Invasiveness , Oncogene Proteins v-erbB/genetics , Oncogene Proteins v-erbB/physiology , Stem Cells/physiology , Transcription Factors/genetics , Transcription Factors/physiology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/physiology
13.
Mech Dev ; 124(9-10): 657-67, 2007.
Article in English | MEDLINE | ID: mdl-17716876

ABSTRACT

ErbB signaling regulates cell adhesion and movements during Xenopus gastrulation, but the downstream pathways involved have not been elucidated. In this study, we show that phosphatidylinositol-3 kinase (PI3K) and Erk mitogen-activated protein kinase (MAPK) mediate ErbB signaling to regulate gastrulation. Both PI3K and MAPK function sequentially in mesoderm specification and movements, and ErbB signaling is important only for the late phase activation of these pathways to control cell behaviors. Activation of either PI3K or Erk MAPK rescues gastrulation defects in ErbB4 morphant embryos, and restores convergent extension in the trunk mesoderm as well as coherent cell migration in the head mesoderm. The two signals preferentially regulate different aspects of cell behaviors, with PI3K more efficient in rescuing cell adhesion and spreading and MAPK more effective in stimulating the formation of filopodia. PI3K and MAPK also weakly activate each other, and together they modulate gastrulation movements. Our results reveal that PI3K and Erk MAPK, which have previously been considered as mesodermal inducing signals, also act downstream of ErbB signaling to participate in regulation of gastrulation morphogenesis.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/physiology , Gastrula/enzymology , Gastrulation/physiology , Oncogene Proteins v-erbB/metabolism , Phosphatidylinositol 3-Kinases/physiology , Signal Transduction/physiology , Animals , Mesoderm/enzymology , Oncogene Proteins v-erbB/physiology , Xenopus laevis/embryology
14.
World J Gastroenterol ; 12(46): 7440-50, 2006 Dec 14.
Article in English | MEDLINE | ID: mdl-17167831

ABSTRACT

Members of the receptor tyrosine kinase family, that include EGFR, ErbB-2/HER-2, ErbB-3/HER-3 and ErbB-4/HER-4, are frequently implicated in experimental models of epithelial cell neoplasia as well as in human cancers. Therefore, interference with the activation of these growth factor receptors represents a promising strategy for development of novel and selective anticancer therapies. Indeed, a number of inhibitors that target either EGFR or HER-2, with the exception of a few that target both; have been developed for treatment of epithelial cancers. Since most solid tumors express different ErbB receptors and/or their ligands, identification of inhibitor(s), targeting multiple EGFR family members may provide a therapeutic benefit to a broader patient population. Here we describe the significance of an ErbB family of receptors in epithelial cancers, and summarize different available therapeutics targeting these receptors. It also emphasizes the need to develop pan-ErbB inhibitors and discusses EGF-Receptor Related Protein, a recently isolated negative regulator of EGFR as a potential pan-ErbB therapeutic for a wide variety of epithelial cancers.


Subject(s)
Gastrointestinal Neoplasms/therapy , Animals , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/physiology , Gastrointestinal Neoplasms/physiopathology , Glycoproteins/pharmacology , Glycoproteins/physiology , Humans , Oncogene Proteins v-erbB/antagonists & inhibitors , Oncogene Proteins v-erbB/physiology , Recombinant Proteins/pharmacology , Signal Transduction
15.
Am J Pathol ; 169(4): 1402-14, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17003495

ABSTRACT

Exposure to ultraviolet (UV) irradiation is the major cause of nonmelanoma skin cancer, the most common form of cancer in the United States. UV irradiation has a variety of effects on the skin associated with carcinogenesis, including DNA damage and effects on signal transduction. The alterations in signaling caused by UV regulate inflammation, cell proliferation, and apoptosis. UV also activates the orphan receptor tyrosine kinase and proto-oncogene Erbb2 (HER2/neu). In this study, we demonstrate that the UV-induced activation of Erbb2 regulates the response of the skin to UV. Inhibition or knockdown of Erbb2 before UV irradiation suppressed cell proliferation, cell survival, and inflammation after UV. In addition, Erbb2 was necessary for the UV-induced expression of numerous proinflammatory genes that are regulated by the transcription factors nuclear factor-kappaB and Comp1, including interleukin-1beta, prostaglandin-endoperoxidase synthase 2 (Cyclooxygenase-2), and multiple chemokines. These results reveal the influence of Erbb2 on the UV response and suggest a role for Erbb2 in UV-induced pathologies such as skin cancer.


Subject(s)
Gene Expression Regulation , Oncogene Proteins v-erbB/physiology , Radiodermatitis/genetics , Skin Neoplasms/etiology , Skin/radiation effects , Ultraviolet Rays , Animals , Apoptosis/genetics , Benzothiazoles/pharmacology , Binding Sites , Cell Proliferation , Chemokines/genetics , Cyclooxygenase 2/genetics , Female , Interleukin-1beta/genetics , Mice , Mice, Inbred Strains , Oncogene Proteins v-erbB/antagonists & inhibitors , Oncogene Proteins v-erbB/genetics , Protein Kinase Inhibitors/pharmacology , Radiodermatitis/metabolism , Skin/metabolism , Skin Neoplasms/genetics , Transcription Factors/metabolism , Transcription, Genetic/radiation effects , Tyrphostins/pharmacology
16.
J Biol Chem ; 280(29): 27383-92, 2005 Jul 22.
Article in English | MEDLINE | ID: mdl-15888451

ABSTRACT

The role of the ErbB family in supporting the malignant phenotype was characterized by stable transfection of a single chain antibody (ScFv5R) against ErbB2 containing a KDEL endoplasmic reticulum retention sequence into GEO human colon carcinoma cells. The antibody traps ErbB2 in the endoplasmic reticulum, thereby down-regulating cell surface ErbB2. The transfected cells showed inactivation of ErbB2 tyrosine phosphorylation and reduced heterodimerization of ErbB2 and ErbB3. This resulted in greater sensitivity to apoptosis induced by growth deprivation and delayed tumorigenicity in vivo. Furthermore, decreased heterodimerization of ErbB2 and ErbB3 led to a reorganization in ErbB function in transfected cells as heterodimerization between epidermal growth factor receptor (EGFR) and ErbB3 increased, whereas ErbB3 activation remained almost the same. Importantly, elimination of ErbB2 signaling resulted in an increase in EGFR expression and activation in transfected cells. Increased EGFR activation contributed to the sustained cell survival in transfected cells.


Subject(s)
Colonic Neoplasms/pathology , Receptor, ErbB-2/physiology , Cell Line, Tumor , Cell Survival , Colonic Neoplasms/etiology , Dimerization , Endoplasmic Reticulum/metabolism , Epidermal Growth Factor/metabolism , Humans , Immunoglobulin Variable Region/pharmacology , Oncogene Proteins v-erbB/physiology , Phosphorylation/drug effects , Protein Transport/drug effects , Receptor, ErbB-2/immunology , Signal Transduction , Transfection
18.
Exp Cell Res ; 296(2): 285-93, 2004 Jun 10.
Article in English | MEDLINE | ID: mdl-15149858

ABSTRACT

The v-ErbB retroviral oncogene is a transduced, mutated copy of the avian EGF receptor gene, and its expression is sufficient to induce tumor formation in vivo. The structural alterations that release the oncogenic potential of the v-ErbB oncogene are similar to EGFR gene mutations described in human tumors. Thus, the study of v-ErbB tumor biology offers a useful model through which we can gain insight into the mechanism of EGFR-induced malignancies. Despite years of study, however, questions remain regarding the domains of v-ErbB required for oncogenicity. We sought to clarify the role of the transmembrane domain of v-ErbB during transformation using S3-v-ErbB, an acutely transforming retroviral oncogene isolated from avian sarcomas. Infection of primary fibroblasts with a retroviral vector containing S3-v-ErbB results in the formation of a transformation-associated phosphoprotein signaling complex, soft agar colony formation, and the rapid induction of highly vascularized sarcomas in vivo. To address contribution of the transmembrane domain of S3-v-ErbB during these processes, we constructed a mutant version of this oncogene with a precise deletion in this domain. Specifically, the S3-v-ErbB-TM- mutant was created through an in-frame deletion of the entire transmembrane domain. Primary fibroblasts expressing this S3-v-ErbB-TM- mutant fail to form a characteristic transformation-associated phosphoprotein complex and do not grow in an anchorage-independent manner. In addition, day-old chicks injected with a helper-independent retrovirus expressing the S3-v-ErbB-TM- mutant exhibit only limited tumor formation in vivo. These results demonstrate that the transmembrane domain and, consequently membrane localization, are essential for S3-v-ErbB-mediated transformation.


Subject(s)
Cell Membrane/metabolism , Cell Transformation, Neoplastic , Oncogene Proteins v-erbB/metabolism , Oncogene Proteins v-erbB/physiology , Animals , Animals, Newborn , Cell Line , Chick Embryo , Chickens , Fibroblasts , Ligands , Mutagenesis, Site-Directed , Mutation , Neoplasms/etiology , Oncogene Proteins v-erbB/administration & dosage , Protein Transport , Transformation, Genetic
19.
Biochem Soc Trans ; 31(Pt 6): 1198-202, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14641025

ABSTRACT

AngII (angiotensin II) and its G-protein-coupled AT(1) receptor play critical roles in mediating cardiovascular diseases such as hypertension, atherosclerosis and restenosis after vascular injury. It is widely believed that AngII promotes these diseases by inducing vascular remodelling that involves hypertrophy, hyperplasia and migration of VSMCs (vascular smooth muscle cells). We have shown that transactivation of an ErbB family receptor, EGFR (epidermal growth factor receptor; ErbB1), is essential for VSMC hypertrophy and migration induced by AngII. However, the precise signal transduction mechanism by which AngII transactivates EGFR/ErbB1 and whether other ErbBs are also required for AngII function remains unclear. Recent studies suggest an involvement of a metalloprotease-dependent ErbB family ligand production in the transactivation. Here, we will discuss the roles and mechanisms of AngII/AT(1) receptor in promoting ErbB receptors transactivation in VSMCs. Further elucidation of this ErbB activation machinery not only will give us a better understanding of the critical molecular mechanism underlying vascular remodelling stimulated by AngII, but will also contribute to development of novel treatment strategies for cardiovascular diseases.


Subject(s)
ErbB Receptors/genetics , Metalloproteases/metabolism , Oncogene Proteins v-erbB/physiology , Transcriptional Activation/physiology , Animals , COS Cells , GTP-Binding Proteins/physiology , Oncogene Proteins v-erbB/metabolism
20.
Exp Cell Res ; 284(1): 111-21, 2003 Mar 10.
Article in English | MEDLINE | ID: mdl-12648470

ABSTRACT

Relay of information from the extracellular environment into the cell often results from a peptide growth factor binding to its cognate cell surface receptor; this event is an integral mechanism by which many cellular functions occur, including cell growth, motility, and survival. In recent years, however, this requirement for ligand binding has been shown to be surpassed by several distinct mechanisms, including cell surface receptor cross-talk (e.g., between epidermal growth factor receptor [EGFR] and G-coupled receptors), receptor-extracellular matrix interactions (e.g., EGFR: integrin complexes), and finally by structural mutations within the receptor itself. While all of these pathways result in so-called ligand-independent signaling by the EGF receptor, to date, only structural mutations in the receptor have been shown to result in qualitative changes in downstream targets of the receptor, which specifically result in oncogenic signaling, transformation, and tumorigenicity. In this review, we describe aspects of the known signaling properties of the retroviral oncogene v-ErbB as a model of ligand-independent oncogenic signaling, and compare these properties to results emerging from ongoing studies on structurally related EGF receptor mutants originally identified in human tumors. A better understanding of the signaling pathways used by these uniquely oncogenic receptor tyrosine kinase mutants may ultimately reveal new targets for the development of novel therapeutics selective for the inhibition of tumor cell growth.


Subject(s)
ErbB Receptors/metabolism , Oncogene Proteins v-erbB/physiology , Signal Transduction/physiology , Animals , Cell Division , Cell Transformation, Neoplastic/metabolism , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic , Humans , Ligands , Mutation , Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL
...