Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Pharmacol Exp Ther ; 345(3): 393-403, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23528611

ABSTRACT

Transactivation of epidermal growth factor receptor (EGFR) signaling by G protein-coupled receptors has been implicated in several cardiovascular (CV) conditions, including hypertension, heart failure, and cardiac and vascular hypertrophy. However, the therapeutic potential of EGFR inhibition in these conditions is currently unknown. The main objective of the present study was to investigate cardiac, vascular, and renal effects of EGFR inhibition by 4-[4-[[(1R)-1-phenylethyl]amino]-7H-pyrrolo[2,3-d]pyrimidin-6-yl]phenol (PKI-166) in the hypertensive chronic kidney disease model. Rats underwent 5/6 nephrectomy (5/6Nx) and were treated with PKI-166, lisinopril or vehicle from week 6 after disease induction until week 12. Sham animals received either PKI-166 or vehicle. Treatment with PKI-166 did not affect the development of the characteristic renal features in 5/6Nx, including proteinuria, diminished creatinine clearance, and increased glomerulosclerosis, whereas these were attenuated by lisinopril. Despite absence of effects on progressive renal damage, PKI-166 attenuated the progression of hypertension and maintained cardiac function (left ventricle end-diastolic pressure) to a similar extent as lisinopril. Also, PKI-166 attenuated the increase in phosphorylated EGFR in the heart as induced by 5/6Nx. Moreover, PKI-166 and lisinopril restored the impaired contraction of isolated thoracic aortic rings to phenylephrine and angiotensin II and impaired myogenic constriction of small mesenteric arteries in 5/6Nx rats. Blockade of the EGFR displays a CV benefit independent of limiting the progression of renal injury. Our findings extend the evidence on EGFR signaling as a target in CV disorders.


Subject(s)
Cardiotonic Agents/pharmacology , ErbB Receptors/antagonists & inhibitors , Hypertension, Renal/drug therapy , Kidney/drug effects , Nephrectomy , Pyrimidines/pharmacology , Pyrroles/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Aorta, Thoracic/drug effects , Blood Pressure/drug effects , Hypertension, Renal/physiopathology , Immunohistochemistry , Lisinopril/pharmacology , Male , Mesenteric Arteries/drug effects , Muscle Contraction/drug effects , Muscle Tonus/drug effects , Muscle, Smooth, Vascular/drug effects , Proteinuria/metabolism , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/drug effects , Receptors, Adrenergic, alpha-1/drug effects
2.
Nephrol Dial Transplant ; 27(5): 2114-22, 2012 May.
Article in English | MEDLINE | ID: mdl-22015440

ABSTRACT

BACKGROUND: Interstitial fibrosis and tubular atrophy (IF/TA) is an important cause of renal function loss and ischaemia-reperfusion (I/R) injury is considered to play an important role in its pathophysiology. The aim of the present study was to investigate the role of a disintegrin and metalloproteinase 17 (ADAM17) in human renal allograft disease and in experimental I/R injury of the kidney. METHODS: We studied the expression of ADAM17 messenger RNA (mRNA) in IF/TA and control kidneys by reverse transcription-polymerase chain reaction and in situ hybridization. Moreover, we assessed ADAM17-mediated heparin-binding epidermal growth factor (HB-EGF) shedding in immortalized human cells. Finally, we studied the effect of pharmacological ADAM17 inhibition in a model of renal I/R injury in rats. RESULTS: ADAM17 mRNA was up-regulated in IF/TA when compared to control kidneys. In normal kidneys, ADAM17 mRNA was weakly expressed in proximal tubules, peritubular capillaries, glomerular endothelium and parietal epithelium. In IF/TA, tubular, capillary and glomerular ADAM17 expression was strongly enhanced with de novo expression in the mesangium. In interstitial fibrotic lesions, we observed co-localization of ADAM17 with HB-EGF protein. In vitro, inhibition of ADAM17 with TNF484 resulted in a dose-dependent reduction of HB-EGF shedding in phorbol 12-myrisate 13-acetate-stimulated cells and non-stimulated cells. In vivo, ADAM17 inhibition significantly reduced the number of glomerular and interstitial macrophages at Day 4 of reperfusion. CONCLUSIONS: In conclusion, HB-EGF co-expresses with ADAM17 in renal interstitial fibrosis, suggesting a potential interaction in IF/TA. Targeting ADAM17 to reduce epidermal growth factor receptor phosphorylation could be a promising way of intervention in human renal disease.


Subject(s)
ADAM Proteins/metabolism , Kidney Transplantation , Kidney/metabolism , Kidney/pathology , Reperfusion Injury/metabolism , Up-Regulation , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/drug effects , ADAM17 Protein , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Atrophy , Cells, Cultured , Child , Child, Preschool , Dose-Response Relationship, Drug , Female , Fibrosis , Heparin-binding EGF-like Growth Factor , Humans , Hydroxamic Acids/pharmacology , In Vitro Techniques , Intercellular Signaling Peptides and Proteins/metabolism , Kidney/drug effects , Male , Middle Aged , Models, Animal , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reperfusion Injury/pathology , Young Adult
3.
J Am Soc Nephrol ; 21(8): 1309-16, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20595681

ABSTRACT

A mutation in pro-EGF causes isolated hypomagnesemia, and monoclonal antibodies targeting the extracellular domain of the EGF receptor (EGFR) affect epithelial Mg(2+) transport. The effect of the EGFR tyrosine kinase inhibitor erlotinib on Mg(2+) homeostasis, however, remains unknown. Here, we injected C57BL/6 mice with erlotinib for 23 days and observed a small but significant decrease in serum Mg(2+) concentrations at days 16 and 23, but the fractional excretion of Mg(2+) remained unchanged after 23 days. Semiquantitative immunohistochemical evaluation did not reveal detectable changes in renal expression of transient receptor potential melastatin 6 (TRPM6) protein, the channel that mediates Mg(2+) reabsorption. Patch clamp analysis in TRPM6-expressing cells demonstrated that 30 muM erlotinib inhibited EGF-induced changes in TRPM6 current density and tyrosine phosphorylation of EGFR; 0.3 muM erlotinib did not have these effects. Furthermore, 30 muM erlotinib inhibited EGF-stimulated increases in the mobile fraction of endomembrane TRPM6 channels. In summary, erlotinib can influence Mg(2+) handling but its effect on the systemic Mg(2+) concentration seems less potent than that observed with antibody-based EGFR inhibitors. These data suggest that typical human dosages of erlotinib are unlikely to severely affect serum Mg(2+) concentrations.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Magnesium/metabolism , Quinazolines/pharmacology , Animals , ErbB Receptors/biosynthesis , Erlotinib Hydrochloride , Mice , Mice, Inbred C57BL , TRPM Cation Channels/drug effects , TRPM Cation Channels/physiology
4.
J Pathol ; 221(2): 183-92, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20225242

ABSTRACT

The epidermal growth factor (EGF) receptor and its ligands are crucially involved in the renal response to ischaemia. We studied the heparin binding-epidermal growth factor (HB-EGF), a major ligand for the EGF receptor, in experimental and human ischaemia/reperfusion injury (IRI). HB-EGF mRNA and protein expression was studied in rat kidneys and cultured human tubular (HK-2) cells that were subjected to IRI and in human donor kidneys during transplantation. The effect of EGF receptor inhibition was investigated in vivo and in vitro. Furthermore, urinary HB-EGF protein excretion was studied after renal transplantation. Finally, HB-EGF KO and WT mice were subjected to IRI to study the role of HB-EGF in renal injury. HB-EGF mRNA was significantly up-regulated in the early phase of IRI in rats, cells, and human donor biopsies. Treatment with PKI-166 reduces macrophage accumulation and interstitial alpha-SMA in the early phase of IRI in rats. In vitro, PKI-166 causes a marked reduction in HB-EGF-induced cellular proliferation. Urinary HB-EGF is increased after transplantation compared with control urines from healthy subjects. HB-EGF KO mice subjected to IRI revealed significantly less morphological damage after IRI, compared with WT mice. We conclude that IRI results in early induction of HB-EGF mRNA and protein in vivo and in vitro. Absence of HB-EGF and inhibition of the EGF receptor in the early phase of IRI has protective effects, suggesting a modulating role for HB-EGF.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Kidney/metabolism , Reperfusion Injury/metabolism , Adult , Aged , Animals , Cells, Cultured , Disease Models, Animal , Epidermal Growth Factor/antagonists & inhibitors , Female , Heparin-binding EGF-like Growth Factor , Humans , Intercellular Signaling Peptides and Proteins/urine , Kidney Transplantation , Male , Mice , Middle Aged , Pyrimidines/pharmacology , Pyrroles/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Up-Regulation , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...