Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
FASEB J ; 20(11): 1936-8, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16877529

ABSTRACT

Endothelin-1 (ET-1), a powerful vasoconstrictor, is involved in vasospastic diseases such as coronary artery disease and subarachnoidal hemorrhage, as well as in renal and cardiovascular fibrotic remodeling. Transactivation of the epidermal growth factor receptor (EGFR) mediates ET-1 signaling in vascular smooth muscle cells (VSMCs) and isolated arteries. Moreover, EGFR is required for a full constrictive response to ET-1. However, the relevant mechanisms mediating EGFR transactivation in response to ET-1 have not been identified. The present study used isolated arteries and VSMCs to investigate the role of the EGFR ligand heparin binding-epidermal growth factor (HB-EGF) in ET-1-induced transactivation of EGFR, intracellular calcium mobilization, and VSMCs contraction. While baseline blood pressures were similar in HB-EGF-deficient and in wild-type littermate mice, the vasoconstrictor actions of ET-1 were attenuated in HB-EGF-/- animals. In isolated mouse carotid artery segments mounted in an arteriograph, ET-1 caused only a weak increase in isovolumetric tone in HB-EGF-deficient vessels, and this effect was mimicked by inhibition of EGFR tyrosine kinase or phosphoinositide 3-kinase (PI3K) in wild-type arteries with or without endothelium, indicating a specific role in VSMCs. EGFR or PI3K inhibitors had no effect on KCl-induced contraction, which was normal in HB-EGF-deficient mice. To confirm that the abnormal responses in HB-EGF-deficient mice were due to impaired EGFR signaling, we studied VSMCs from waved-2 (wa2) mice; these animals have a mutation causing a partial loss of function of EGFR tyrosine kinase activity. The ET-1-induced calcium peak was reduced by 30% in VSMCs from wa2 mice and from HB-EGF-/- mice. This effect was reproduced by preincubation of wild-type VSMCs with EGFR inhibitor AG1478 and PI3K inhibitors LY294002 and wortmannin. ProHB-EGF is bound to the cell membrane and released after cleavage by metalloproteinases; its action may contribute to effects of GPCR agonists on cell growth. Pretreatment of mouse VSMCs with batimastat, a metalloproteinase inhibitor, significantly attenuated ET-1-induced [Ca(2+)](i) response in wild-type cells. Human proHB-EGF has been shown to be the endogenous receptor for Corynebacterium diphteriae toxin (DT). Mutated DT toxin (CRM197) is devoid of toxicity but it neutralizes HB-EGF binding to EGFR. Pretreatment of human VSMCs from internal mammary arteries with CRM197 significantly blunted ET-1-stimulated calcium transients. In conclusion, these findings suggest that the mechanism of ET-1-induced vasoconstriction involves HB-EGF-mediated transactivation of the EGFR. This functional cascade requires modulation of agonist-induced calcium transient by EGFR and PI3K with extremely fast kinetics, suggesting a novel paradigm for GPCR-mediated calcium signaling, which may offer future therapeutic targets.


Subject(s)
Carotid Arteries/physiology , Endothelin-1/pharmacology , ErbB Receptors/physiology , Heparin/physiology , Animals , Blood Pressure , Carotid Arteries/drug effects , ErbB Receptors/deficiency , ErbB Receptors/genetics , Heparin/deficiency , Heparin/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Point Mutation , Potassium Chloride/pharmacology , Protein Binding
2.
Circ Res ; 95(10): 989-97, 2004 Nov 12.
Article in English | MEDLINE | ID: mdl-15486316

ABSTRACT

Stimulation of alpha1-adrenoceptors induces proliferation of vascular smooth muscle cells (SMCs) and contributes to arterial remodeling. Although activation of NAD(P)H oxidase and generation of reactive oxygen species (ROS) are required, little is known about this pathway. In this study, we examined the hypothesis that epidermal growth factor receptor (EGFR) transactivation and extracellular regulated kinases (ERK) are involved in alpha1-adrenoceptor-mediated SMC growth. Phenylephrine increased protein synthesis in association with a rapid (< or =5 minutes) and sustained (> or =60 minutes) doubling of phosphorylation of EGFR and ERK1/2, but not p38 or JNK in the media of rat aorta maintained in organ culture. Antagonists of EGFR phosphotyrosine activity (AG-1478) and ERK phosphorylation (PD-98059, U-0126) abolished phenylephrine-induced protein synthesis, whereas antagonists of p38 or JNK phosphorylation had no specific effect. A competitive antagonist (P22) for heparin binding EGF-like growth factor (HB-EGF) blocked phenylephrine-induced protein synthesis, as did downregulation of pro-HB-EGF (CRM197). Phenylephrine-induced protein synthesis was inhibited by neutralizing antibody to HB-EGF and absent in HB-EGF-/- SMCs. Inhibitors of metalloproteinases (BiPS, KB-R7785) also blocked adrenergic growth. The neutralizing antibody against HB-EGF had no effect on the two-fold increase in ROS generation induced by phenylephrine (DCF fluorescence), suggesting that stimulation of NAD(P)H oxidase by alpha1-adrenoceptor occupation precedes HB-EGF release. Cell culture studies confirmed and extended these findings. These data suggest that alpha1-adrenoceptor-mediated SMC growth requires ROS-dependent shedding of HB-EGF, transactivation of EGFR, and activation of the MEK1/2-dependent MAP kinase pathway. This trophic pathway may link sympathetic activity to arterial wall growth in adaptive remodeling and hypertrophic disease.


Subject(s)
ErbB Receptors/physiology , Glycine/analogs & derivatives , MAP Kinase Signaling System/physiology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Receptors, Adrenergic, alpha-1/physiology , Adrenergic alpha-1 Receptor Agonists , Animals , Anthracenes/pharmacology , Aorta, Thoracic/injuries , Aorta, Thoracic/pathology , Bacterial Proteins/pharmacology , Benzopyrans/pharmacology , Butadienes/pharmacology , Catheterization/adverse effects , Cell Division , Dipeptides/pharmacology , ErbB Receptors/drug effects , Flavonoids/pharmacology , Glycine/pharmacology , Hydroxamic Acids/pharmacology , Imidazoles/pharmacology , MAP Kinase Kinase 1/physiology , MAP Kinase Kinase 2/physiology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/enzymology , Nitriles/pharmacology , Organ Culture Techniques , Phenylephrine/pharmacology , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Pyridines/pharmacology , Quinazolines , Rats , Thrombin/pharmacology , Tyrphostins/pharmacology
3.
J Mol Med (Berl) ; 81(7): 392-403, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12827270

ABSTRACT

It is becoming clear that converging pathways coordinate early heart valve development and remodeling into functional valve leaflets. The integration of these pathways begins with macro and molecular interactions outside the cell in the extracellular matrix separating the myocardial and endocardial tissue components of the rudimentary heart. Such interactions regulate events at the cell surface through receptors, proteases, and other membrane molecules which in turn transduce signals into the cell. These signals trigger intracellular cascades that transduce cellular responses through both transcription factor and cofactor activation mediating gene induction or suppression. Chamber septation and valve formation occur from these coordinated molecular events within the endocardial cushions to sustain unidirectional blood flow and embryo viability. This review discusses the emerging connection between extracellular matrix and growth factor receptor signaling during endocardial cushion morphogenesis by highlighting the extracellular component, hyaluronan, and erbB receptor functions during early valve development.


Subject(s)
Extracellular Matrix/physiology , Growth Substances/physiology , Heart Valves/embryology , Hyaluronic Acid/physiology , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Animals , Endocardium/embryology , Gene Expression Regulation, Developmental , Growth Substances/genetics , Heart/embryology , Humans , Morphogenesis , Transcriptional Activation
4.
Article in English | MEDLINE | ID: mdl-15649768

ABSTRACT

The effects of ovine prolactin (oPRL) and striped bass prolactin (sbPRL; Morone saxatilis) on plasma osmolality, electrolyte balance, and gill Na(+),K(+)-ATPase activity were investigated in hypophysectomized (Hx), freshwater (FW)-acclimated, hybrid striped bass (M. saxatilisxMorone chrysops). They were kept in dilute (isoosmotic) seawater for about 10 days after surgery. Seven days after transfer to FW, Hx fish had lower plasma osmolality and lower levels of Na(+), Cl(-), and Ca(2+) than sham-operated and intact fish. Fish were injected four times with oPRL (1, 5, or 20 microg/g body mass), sbPRL (10 or 100 ng/g), or hormone vehicle (0.9% NaCl) at 48-h intervals (days 0, 2, 4, and 6) in FW and then sampled for blood plasma 24 h after the fourth injection (day 7). In Hx fish, oPRL (5 and 20 microg/g) and sbPRL (10 and 100 ng/g) were effective in maintaining plasma osmolality and levels of Na(+), Cl(-), and Ca(2+) above values seen in saline-injected controls. Hypophysectomy did not affect branchial Na(+),K(+)-ATPase activity, but enzyme activity was significantly reduced in Hx fish receiving oPRL (20 mug/g) or sbPRL (10 or 100 ng/g). These results indicate that PRL acts to maintain plasma osmotic and ionic balance in FW-adapted hybrid striped bass, and that this may involve downregulation of branchial Na(+),K(+)-ATPase activity.


Subject(s)
Bass/physiology , Hypophysectomy , Prolactin/pharmacology , Adenosine Triphosphatases/metabolism , Animals , Bass/blood , Bass/metabolism , Branchial Region/drug effects , Branchial Region/enzymology , Electrolytes/blood , Injections , Osmolar Concentration , Prolactin/administration & dosage , Water-Electrolyte Balance/drug effects
5.
Ann N Y Acad Sci ; 995: 22-38, 2003 May.
Article in English | MEDLINE | ID: mdl-12814936

ABSTRACT

EGF family growth factors, including transforming growth factor-alpha (TGFalpha), amphiregulin (AR), and heparin-binding EGF (HB-EGF), are invariably expressed as transmembrane precursors that are cleaved at one or more sites in the extracellular domain to release soluble growth factor. Considerable attention has focused on the identification of proteases responsible for these processing events. We previously implicated tumor necrosis factor-alpha converting enzyme (TACE/ADAM17) in the generation of soluble TGFalpha from its transmembrane precursor, proTGFalpha. Here, we review our findings that primary keratinocytes from Tace(deltaZn/deltaZn) mice, which express a nonfunctional TACE, released dramatically lower levels of soluble TGFalpha compared to their normal counterparts, even though TGFalpha mRNA and cell-associated protein levels were similar in the two cell populations. Restoration of TACE activity in Tace(deltaZn/deltaZn) cells increased shedding of TGFalpha species, including the mature, 6-kDa protein. Further, exogenous TACE enzyme accurately cleaved the N-terminal processing site of proTGFalpha in cell lysates, as well as both physiologic sites of a soluble proTGFalpha ectodomain. TACE also accurately cleaved peptide substrates corresponding to the processing sites of several additional EGF family members, and restoration of TACE activity enhanced the shedding of soluble AR and HB-EGF proteins from Tace(deltaZn/deltaZn) cells. Finally, reduction of functional TACE gene dosage greatly exacerbated the open-eye defect of Egfr(wa-2/wa-2) newborns, which is regulated by redundant actions of several EGF family ligands. The implications of these results for the biology of the EGF family and TACE are discussed.


Subject(s)
Epidermal Growth Factor/metabolism , Metalloendopeptidases/physiology , Protein Precursors/metabolism , Transforming Growth Factor alpha/metabolism , ADAM Proteins , ADAM17 Protein , Amino Acid Sequence , Amphiregulin , Animals , EGF Family of Proteins , Epidermal Growth Factor/chemistry , ErbB Receptors/metabolism , Glycoproteins/chemistry , Glycoproteins/metabolism , Heparin-binding EGF-like Growth Factor , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/metabolism , Ligands , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Mice , Mice, Knockout , Protein Precursors/chemistry , Transforming Growth Factor alpha/chemistry
6.
J Biol Chem ; 280(4): 2562-8, 2005 Jan 28.
Article in English | MEDLINE | ID: mdl-15536070

ABSTRACT

The mechanisms that regulate the transition between the initial priming phase and DNA replication in liver regeneration are poorly understood. To study this transition, we compared events occurring after standard two-thirds partial hepatectomy, which elicits full regeneration, with response to a reduced hepatectomy, one-third partial hepatectomy (1/3PH), which leads to little DNA replication. Although the initial response to partial hepatectomy at the priming phase appeared to be similar between the two procedures, cell cycle progression was significantly blunted in 1/3PH mice. Among the main defects observed in 1/3PH mice were an almost complete deficiency in retinoblastoma phosphorylation and the lack of increase in kinase activity associated with cyclin E. We report that, in two-thirds partial hepatectomy mice, the expression of heparin-binding epidermal growth factor-like growth factor (HB-EGF) preceded the start of DNA replication and was not detectable in 1/3PH animals. Injection of HB-EGF into 1/3PH mice resulted in a >15-fold increase in DNA replication. Moreover, we show that hepatocyte DNA replication was delayed in HB-EGF knock-out mice. In summary, we show that HB-EGF is a key factor for hepatocyte progression through G(1)/S transition during liver regeneration.


Subject(s)
Epidermal Growth Factor/metabolism , Heparin/metabolism , Hepatocytes/metabolism , Liver Regeneration , Liver/physiology , Animals , Blotting, Western , Bromodeoxyuridine/pharmacology , Cell Cycle , Cell Proliferation , Cyclin E/metabolism , DNA/metabolism , DNA Replication , G1 Phase , Heparin-binding EGF-like Growth Factor , Hepatocytes/physiology , Immunohistochemistry , Immunoprecipitation , Intercellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , S Phase , Time Factors
7.
EMBO J ; 22(11): 2704-16, 2003 Jun 02.
Article in English | MEDLINE | ID: mdl-12773386

ABSTRACT

Heparin-binding epidermal growth factor (HB-EGF) and betacellulin (BTC) are activating ligands for EGF receptor (EGFR/ErbB1) and ErbB4. To identify their physiological functions, we disrupted mouse HB-EGF and BTC alleles by homologous recombination. Most HB-EGF(-/-) mice died before weaning, and survivors had enlarged, dysfunctional hearts and reduced lifespans. Although BTC(-/-) mice were viable and fertile and displayed no overt defects, the lifespan of double null HB-EGF(-/-)/BTC(-/-) mice was further reduced, apparently due to accelerated heart failure. HB-EGF(-/-) newborns had enlarged and malformed semilunar and atrioventricular heart valves, and hypoplastic, poorly differentiated lungs. Defective cardiac valvulogenesis was the result of abnormal mesenchymal cell proliferation during remodeling, and was associated with dramatic increases in activated Smad1/5/8. Consistent with the phenotype, HB-EGF transcripts were localized to endocardial cells lining the margins of wild-type valves. Similarly defective valvulogenesis was observed in newborn mice lacking EGFR and tumor necrosis factor-alpha converting enzyme (TACE). These results suggest that cardiac valvulogenesis is dependent on EGFR activation by TACE-derived soluble HB-EGF, and that EGFR signaling is required to regulate bone morphogenetic protein signaling in this context.


Subject(s)
Epidermal Growth Factor/deficiency , Heart Valves/abnormalities , Metalloendopeptidases/deficiency , ADAM Proteins , ADAM17 Protein , Animals , Betacellulin , Bone Morphogenetic Proteins/physiology , Epidermal Growth Factor/genetics , Epidermal Growth Factor/physiology , ErbB Receptors/metabolism , Gene Expression Regulation, Developmental , Heart Defects, Congenital/embryology , Heart Defects, Congenital/genetics , Heart Defects, Congenital/metabolism , Heart Valves/embryology , Heart Valves/metabolism , Heparin-binding EGF-like Growth Factor , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/physiology , Lung/abnormalities , Metalloendopeptidases/genetics , Metalloendopeptidases/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, ErbB-4
SELECTION OF CITATIONS
SEARCH DETAIL