Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Mol Life Sci ; 81(1): 272, 2024 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-38900158

RESUMO

We addressed the heteromerization of the epidermal growth factor receptor (EGFR) with G-protein coupled receptors (GPCR) on the basis of angiotensin-II-receptor-subtype-1(AT1R)-EGFR interaction as proof-of-concept and show its functional relevance during synergistic nuclear information transfer, beyond ligand-dependent EGFR transactivation. Following in silico modelling, we generated EGFR-interaction deficient AT1R-mutants and compared them to AT1R-wildtype. Receptor interaction was assessed by co-immunoprecipitation (CoIP), Förster resonance energy transfer (FRET) and fluorescence-lifetime imaging microscopy (FLIM). Changes in cell morphology, ERK1/2-phosphorylation (ppERK1/2), serum response factor (SRF)-activation and cFOS protein expression were determined by digital high content microscopy at the single cell level. FRET, FLIM and CoIP confirmed the physical interaction of AT1R-wildtype with EGFR that was strongly reduced for the AT1R-mutants. Responsiveness of cells transfected with AT1R-WT or -mutants to angiotensin II or EGF was similar regarding changes in cell circularity, ppERK1/2 (direct and by ligand-dependent EGFR-transactivation), cFOS-expression and SRF-activity. By contrast, the EGFR-AT1R-synergism regarding these parameters was completely absent for in the interaction-deficient AT1R mutants. The results show that AT1R-EGFR heteromerisation enables AT1R-EGFR-synergism on downstream gene expression regulation, modulating the intensity and the temporal pattern of nuclear AT1R/EGFR-information transfer. Furthermore, remote EGFR transactivation, via ligand release or cytosolic tyrosine kinases, is not sufficient for the complete synergistic control of gene expression.


Assuntos
Núcleo Celular , Receptores ErbB , Receptor Tipo 1 de Angiotensina , Receptores ErbB/metabolismo , Humanos , Receptor Tipo 1 de Angiotensina/metabolismo , Receptor Tipo 1 de Angiotensina/genética , Núcleo Celular/metabolismo , Transferência Ressonante de Energia de Fluorescência , Fosforilação , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Membrana Celular/metabolismo , Angiotensina II/metabolismo , Angiotensina II/farmacologia , Fator de Crescimento Epidérmico/metabolismo , Células HEK293 , Ligação Proteica , Fator de Resposta Sérica/metabolismo , Fator de Resposta Sérica/genética
2.
Sci Rep ; 13(1): 22827, 2023 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-38129563

RESUMO

Endothelial cells (EC) are key players in vascular function, homeostasis and inflammation. EC show substantial heterogeneity due to inter-individual variability (e.g. sex-differences) and intra-individual differences as they originate from different organs or vessels. This variability may lead to different responsiveness to external stimuli. Here we compared the responsiveness of female human primary EC from the aorta (HAoEC) and coronary arteries (HCAEC) to Epidermal Growth Factor Receptor (EGFR) activation. EGFR is an important signal integration hub for vascular active substances with physiological and pathophysiological relevance. Our transcriptomic analysis suggested that EGFR activation differentially affects the inflammatory profiles of HAoEC and HCAEC, particularly by inducing a HCAEC-driven leukocyte attraction but a downregulation of adhesion molecule and chemoattractant expression in HAoEC. Experimental assessments of selected inflammation markers were performed to validate these predictions and the results confirmed a dual role of EGFR in these cells: its activation initiated an anti-inflammatory response in HAoEC but a pro-inflammatory one in HCAEC. Our study highlights that, although they are both arterial EC, female HAoEC and HCAEC are distinguishable with regard to the role of EGFR and its involvement in inflammation regulation, what may be relevant for vascular maintenance but also the pathogenesis of endothelial dysfunction.


Assuntos
Vasos Coronários , Células Endoteliais , Humanos , Feminino , Vasos Coronários/metabolismo , Células Endoteliais/metabolismo , Células Cultivadas , Aorta , Receptores ErbB/metabolismo , Inflamação/metabolismo , Endotélio Vascular/metabolismo
3.
Cell Mol Life Sci ; 79(1): 57, 2021 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-34921637

RESUMO

The tyrosine kinase receptor EGFR and the G-protein-coupled receptor AT1R induce essential cellular responses, in part via receptor crosstalk with an unknown role in nuclear information transfer and transcription regulation. We investigated whether this crosstalk results in linear, EGFR-mediated nuclear signalling or in parallel, synergistic information transfer leading to qualitative and temporal variations, relevant for gene expression and environment interaction. AT1R and EGFR synergistically activate SRF via the ERK1/2-TCF and actin-MRTF pathways. Synergism, comprised of switch-like and graded single cell response, converges on the transcription factors AP1 and EGR, resulting in synergistic transcriptome alterations, in qualitative (over-additive number of genes), quantitative (over-additive expression changes of individual genes) and temporal (more late onset and prolonged expressed genes) terms. Gene ontology and IPA® pathway analysis indicate prolonged cell stress (e.g. hypoxia-like) and dysregulated vascular biology. Synergism occurs during separate but simultaneous activation of both receptors and during AT1R-induced EGFR transactivation. EGFR and AT1R synergistically regulate gene expression in qualitative, quantitative and temporal terms with (patho)physiological relevance, extending the importance of EGFR-AT1R crosstalk beyond cytoplasmic signalling.


Assuntos
Regulação da Expressão Gênica , Receptor Tipo 1 de Angiotensina/metabolismo , Receptores ErbB/metabolismo , Células HEK293 , Humanos , Receptor Cross-Talk , Transdução de Sinais , Transcriptoma
4.
Sci Rep ; 11(1): 7269, 2021 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-33790318

RESUMO

Vascular EGF receptors (EGFR) influence function and structure of arterial vessels. In genetic mouse models we described the role of vascular smooth muscle (VSMC) EGFR for proper physiological function and structure as well as for pathophysiological alterations by obesity or angiotensin II. As the importance of endothelial (EC) EGFR in vivo is unknown, we analyzed the impact of EC-EGFR knockout in a conditional mouse model on vascular and renal function under control condition as well as in obesity and in comparison to VSMC-KO. Heart and lung weight, blood pressure and aortic transcriptome (determined by RNA-seq) were not affected by EC-EGFR-KO. Aortic reactivity to α1-adrenergic stimulation was not affected by EC-EGFR-KO contrary to VSMC-EGFR-KO. Endothelial-induced relaxation was reduced in abdominal aorta of EC-EGFR-KO animals, whereas it was enhanced in VSMC-EGFR-KO animals. Mesenteric arteries of EC-EGFR-KO animals showed enhanced sensitivity to α1-adrenergic stimulation, whereas endothelial-induced relaxation and vessel morphology were not affected. Renal weight, histomorphology, function (albumin excretion, serum creatinine, fractional water excretion) or transcriptome were not affected by EC-EGFR-KO, likewise in VSMC-EGFR-KO. High fat diet (HFD) over 18 weeks induced arterial wall thickening, renal weight increase, creatininemia, renal and aortic transcriptome alterations with a similar pattern in EC-EGFR-WT and EC-EGFR-KO animals by contrast to the previously reported impact of VSMC-EGFR-KO. HFD induced endothelial dysfunction in abdominal aortae of EC-EGFR-WT, which was not additive to the EC-EGFR-KO-induced endothelial dysfunction. As shown before, VSMC-EGFR-KO prevented HFD-induced endothelial dysfunction. HFD-induced albuminuria was less pronounced in EC-EGFR-KO animals and abrogated in VSMC-EGFR-KO animals. Our results indicate that EC-EGFR, in comparison to VSMC-EGFR, is of minor and opposite importance for basal renovascular function as well as for high fat diet-induced vascular remodeling and renal end organ damage.


Assuntos
Aorta Abdominal/metabolismo , Dieta Hiperlipídica/efeitos adversos , Endotélio Vascular/metabolismo , Receptores ErbB/metabolismo , Rim/metabolismo , Obesidade/metabolismo , Animais , Receptores ErbB/genética , Camundongos , Camundongos Knockout , Obesidade/induzido quimicamente , Obesidade/genética
5.
Diabetologia ; 63(10): 2218-2234, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32548701

RESUMO

AIMS/HYPOTHESIS: Obesity causes type 2 diabetes leading to vascular dysfunction and finally renal end-organ damage. Vascular smooth muscle (VSM) EGF receptor (EGFR) modulates vascular wall homeostasis in part via serum response factor (SRF), a major regulator of VSM differentiation and a sensor for glucose. We investigated the role of VSM-EGFR during obesity-induced renovascular dysfunction, as well as EGFR-hyperglycaemia crosstalk. METHODS: The role of VSM-EGFR during high-fat diet (HFD)-induced type 2 diabetes was investigated in a mouse model with inducible, VSM-specific EGFR-knockout (KO). Various structural and functional variables as well as transcriptome changes, in vivo and ex vivo, were assessed. The impact of hyperglycaemia on EGFR-induced signalling and SRF transcriptional activity and the underlying mechanisms were investigated at the cellular level. RESULTS: We show that VSM-EGFR mediates obesity/type 2 diabetes-induced vascular dysfunction, remodelling and transcriptome dysregulation preceding renal damage and identify an EGFR-glucose synergism in terms of SRF activation, matrix dysregulation and mitochondrial function. EGFR deletion protects the animals from HFD-induced endothelial dysfunction, creatininaemia and albuminuria. Furthermore, we show that HFD leads to marked changes of the aortic transcriptome in wild-type but not in KO animals, indicative of EGFR-dependent SRF activation, matrix dysregulation and mitochondrial dysfunction, the latter confirmed at the cellular level. Studies at the cellular level revealed that high glucose potentiated EGFR/EGF receptor 2 (ErbB2)-induced stimulation of SRF activity, enhancing the graded signalling responses to EGF, via the EGFR/ErbB2-ROCK-actin-MRTF pathway and promoted mitochondrial dysfunction. CONCLUSIONS/INTERPRETATION: VSM-EGFR contributes to HFD-induced vascular and subsequent renal alterations. We propose that a potentiated EGFR/ErbB2-ROCK-MRTF-SRF signalling axis and mitochondrial dysfunction underlie the role of EGFR. This advanced working hypothesis will be investigated in mechanistic depth in future studies. VSM-EGFR may be a therapeutic target in cases of type 2 diabetes-induced renovascular disease. DATA AVAILABILITY: The datasets generated during and/or analysed during the current study are available in: (1) share_it, the data repository of the academic libraries of Saxony-Anhalt ( https://doi.org/10.25673/32049.2 ); and (2) in the gene expression omnibus database with the study identity GSE144838 ( https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE144838 ). Graphical abstract.


Assuntos
Aorta/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Angiopatias Diabéticas/genética , Nefropatias Diabéticas/genética , Receptores ErbB/genética , Músculo Liso Vascular/metabolismo , Obesidade/metabolismo , Fator de Resposta Sérica/metabolismo , Actinas/metabolismo , Animais , Aorta/fisiopatologia , Linhagem Celular , Diabetes Mellitus Tipo 2/fisiopatologia , Angiopatias Diabéticas/metabolismo , Angiopatias Diabéticas/fisiopatologia , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/fisiopatologia , Dieta Hiperlipídica , Células HEK293 , Humanos , Hiperglicemia/metabolismo , Hiperglicemia/fisiopatologia , Camundongos , Camundongos Knockout , Músculo Liso Vascular/fisiopatologia , Miócitos de Músculo Liso , Obesidade/fisiopatologia , Transdução de Sinais , Remodelação Vascular , Quinases Associadas a rho/metabolismo
6.
Br J Pharmacol ; 175(14): 2956-2967, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29682743

RESUMO

BACKGROUND AND PURPOSE: The mineralocorticoid receptor (MR) contributes to fibrosis in various tissues, and MR antagonists, like eplerenone, are used to prevent fibrosis. The role of MR antagonists in hepatic fibrosis and cirrhosis is unknown. Here, we investigated the role of MRs and eplerenone in cirrhosis development. EXPERIMENTAL APPROACH: Liver fibrosis (5 weeks) and cirrhosis, without (8 weeks) and with ascites (12 weeks), were induced by CCl4 in rats and comprehensively analysed. The effect of eplerenone on the development of cirrhosis with ascites was assessed. MR expression, cellular and subcellular distribution and impact of hypoxia were investigated in vivo and ex vivo. Primary rat hepatocytes and cell lines were used to investigate MR trafficking and transcriptional activity mechanistically. KEY RESULTS: In cirrhosis with ascites, MR mRNA and protein expressions were reduced in hepatocytes of hypoxic areas. While in normoxic areas MRs were mainly cytosolic, the remaining MRs in hypoxic areas were mainly localized in the nuclei, indicating activation followed by translocation and degradation. Accordingly, eplerenone treatment prevented nuclear MR translocation and the worsening of cirrhosis. Exposing hepatocytes ex vivo to hypoxia induced nuclear MR translocation and enhanced transcriptional MR activity at response elements of the NF-κB pathway. CONCLUSIONS AND IMPLICATIONS: We showed for the first time that hypoxia leads to a pathogenetic ligand-independent activation of hepatic MRs during cirrhosis resulting in their nuclear translocation and transcriptional activation of the NF-κB pathway. Treatment with eplerenone prevented the worsening of cirrhosis by blocking this ligand-independent activation of the MR.


Assuntos
Eplerenona/uso terapêutico , Cirrose Hepática/tratamento farmacológico , Antagonistas de Receptores de Mineralocorticoides/uso terapêutico , Receptores de Mineralocorticoides/fisiologia , Animais , Tetracloreto de Carbono , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Eplerenona/farmacologia , Células HEK293 , Hepatócitos/metabolismo , Humanos , Fígado/metabolismo , Fígado/patologia , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Masculino , Antagonistas de Receptores de Mineralocorticoides/farmacologia , RNA Mensageiro/metabolismo , Ratos Wistar , Receptores de Mineralocorticoides/genética
7.
Biochim Biophys Acta ; 1863(7 Pt A): 1519-33, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27012600

RESUMO

Besides their importance for the vascular tone, vascular smooth muscle cells (VSMC) also contribute to pathophysiological vessel alterations. Various G-protein coupled receptor ligands involved in vascular dysfunction and remodeling can transactivate the epidermal growth factor receptor (EGFR) of VSMC, yet the importance of EGFR transactivation for the VSMC phenotype is incompletely understood. The aims of this study were (i) to characterize further the importance of the VSMC-EGFR for proliferation, migration and marker gene expression for inflammation, fibrosis and reactive oxygen species (ROS) homeostasis and (ii) to test the hypothesis that vasoactive substances (endothelin-1, phenylephrine, thrombin, vasopressin and ATP) rely differentially on the EGFR with respect to the abovementioned phenotypic alterations. In primary, aortic VSMC from mice without conditional deletion of the EGFR, proliferation, migration, marker gene expression (inflammation, fibrosis and ROS homeostasis) and cell signaling (ERK 1/2, intracellular calcium) were analyzed. VSMC-EGFR loss reduced collective cell migration and single cell migration probability, while no difference between the genotypes in single cell velocity, chemotaxis or marker gene expression could be observed under control conditions. EGF promoted proliferation, collective cell migration, chemokinesis and chemotaxis and leads to a proinflammatory gene expression profile in wildtype but not in knockout VSMC. Comparing the impact of five vasoactive substances (all reported to transactivate EGFR and all leading to an EGFR dependent increase in ERK1/2 phosphorylation), we demonstrate that the importance of EGFR for their action is substance-dependent and most apparent for crowd migration but plays a minor role for gene expression regulation.


Assuntos
Movimento Celular , Receptores ErbB/metabolismo , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/enzimologia , Trifosfato de Adenosina/farmacologia , Animais , Movimento Celular/efeitos dos fármacos , Proliferação de Células , Células Cultivadas , Endotelina-1/farmacologia , Ativação Enzimática , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/agonistas , Receptores ErbB/deficiência , Receptores ErbB/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibrose , Regulação da Expressão Gênica , Genótipo , Inflamação/genética , Inflamação/metabolismo , Ligantes , Camundongos Knockout , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Estresse Oxidativo , Fenótipo , Fenilefrina/farmacologia , Cultura Primária de Células , Transdução de Sinais , Trombina/farmacologia , Fatores de Tempo , Vasopressinas/farmacologia
8.
Clin Sci (Lond) ; 130(1): 19-33, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26438881

RESUMO

Epi dermal growth factor (EGF) receptor (EGFR) is activated by its canonical ligands and transactivated by various vasoactive substances, e.g. angiotensin II (Ang II). Vascular EGFR has been proposed to be involved in vascular tissue homoeostasis and remodelling. Thus, most studies have focused on its role during long-term vascular changes whereas the relevance for acute regulation of vascular function in vivo and ex vivo is insufficiently understood. To investigate the postnatal role of VSMCs (vascular smooth muscle cells) EGFR in vivo and ex vivo, we generated a mouse model with cell-specific and inducible deletion of VSMC EGFR and studied the effect on basal blood pressure, acute pressure response to, among others, Ang II in vivo as well as ex vivo, cardiovascular tissue homoeostasis and vessel morphometry in male mice. In knockout (KO) animals, systolic, diastolic and mean blood pressures were reduced compared with wild-type (WT). Furthermore, Ang II-induced pressure load was lower in KO animals, as was Ang II-induced force development and extracellular-signal-regulated kinase 1 and 2 (ERK1/2) phosphorylation in aortic rings from KO animals. By contrast, we observed no difference in force development during application of serotonin, KCl, endothelin-1 or endothelin-1-induced pressure load in KO animals. In addition, nitric oxide (NO)-mediated vasodilation was not affected. Heart weight (HW) increase and up-regulation of aortic and cardiac expression of Ccl2 (chemoattractant protein-2) and serpinE1 (plasminogen activator inhibitor 1) during the transition from 4- to 10-months of age were prevented by VSMC EGFR KO. We conclude that VSMC EGFR is involved in basal blood pressure homoeostasis and acute pressure response to Ang II, and thereby contributes to maturation-related remodelling.


Assuntos
Angiotensina II , Pressão Sanguínea , Receptores ErbB/deficiência , Hipertensão/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Fatores Etários , Animais , Pressão Sanguínea/efeitos dos fármacos , Cardiomegalia/induzido quimicamente , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatologia , Quimiocina CCL2/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Receptores ErbB/genética , Deleção de Genes , Humanos , Hipertensão/induzido quimicamente , Hipertensão/genética , Hipertensão/fisiopatologia , Hipertensão/prevenção & controle , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/fisiopatologia , Miócitos de Músculo Liso/efeitos dos fármacos , Fosforilação , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Transdução de Sinais , Fatores de Tempo , Remodelação Vascular , Vasoconstritores/farmacologia
9.
Sci Rep ; 4: 7430, 2014 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-25503263

RESUMO

Non-physiological activation of the mineralocorticoid receptor (MR), e.g. by aldosterone under conditions of high salt intake, contributes to the pathogenesis of cardiovascular diseases, although beneficial effects of aldosterone also have been described. The epidermal growth factor receptor (EGFR) contributes to cardiovascular alterations and mediates part of the MR effects. Recently, we showed that EGFR is required for physiological homeostasis and function of heart and arteries in adult animals. We hypothesize that moderate high aldosterone/NaCl, at normal blood pressure, affects the cardiovascular system depending on cardiovascular EGFR. Therefore we performed an experimental series in male and female animals each, using a recently established mouse model with EGFR knockout in vascular smooth muscle cells and cardiomyocytes and determined the effects of a mild-high aldosterone-to-NaCl constellation on a.o. marker gene expression, heart size, systolic blood pressure, impulse conduction and heart rate. Our data show that (i) cardiac tissue of male but not of female mice is sensitive to mild aldosterone/NaCl treatment, (ii) EGFR knockout induces stronger cardiac disturbances in male as compared to female animals and (iii) mild aldosterone/NaCl treatment requires the EGFR in order to disturb cardiac tissue homeostasis whereas beneficial effects of aldosterone seem to be independent of EGFR.


Assuntos
Aldosterona/fisiologia , Receptores ErbB/fisiologia , Miocárdio/metabolismo , Cloreto de Sódio/metabolismo , Aldosterona/farmacologia , Animais , Biomarcadores/metabolismo , Feminino , Homeostase , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Cloreto de Sódio/farmacologia , Transcriptoma
10.
Cell Physiol Biochem ; 33(4): 1106-16, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24732969

RESUMO

BACKGROUND/AIMS: Chronic renal proximal tubule dysfunction after therapy with the antineoplastic agent ifosfamide (IFO) is often attributed to the metabolite chloroacetaldehyde (CAA). Chronic IFO-nephropathy is reported to result in tubulointerstitial fibrosis and inflammation. METHODS: To elucidate possible effects of CAA on extracellular matrix homeostasis, we investigated the action of CAA on markers of extracellular matrix (ECM) homeostasis in human proximal tubule cells (RPTEC) by use of direct ELISA for extracellular collagens and gelatin zymography. RESULTS: An increase in type III collagen and a decrease in type IV collagen abundance in the media of RPTEC could be observed after exposure to CAA in clinically relevant concentrations. CAA increased intracellular type III and decreased intracellular type IV collagen. MMP-2 activity was decreased but MMP-9 activity unchanged. The enhanced CAA-induced collagen III formation could be attenuated by the intracellular Ca(2+)-chelator BAPTA-AM, the PKA-antagonist H-89 and by extracellular acidification. CAA-induced collagen III abundance was enhanced by db-cAMP and IBMX and by protein overload. CONCLUSIONS: CAA exerts profibrotic effects on RPTEC dependent on Ca(2+) and cAMP/PKA-signaling. These effects are enhanced by additional protein burden and attenuated by acidification. © 2014 S. Karger AG, Basel.


Assuntos
Acetaldeído/análogos & derivados , Matriz Extracelular/metabolismo , Túbulos Renais Proximais/efeitos dos fármacos , Acetaldeído/farmacologia , Antineoplásicos Alquilantes/química , Antineoplásicos Alquilantes/metabolismo , Cálcio/metabolismo , Células Cultivadas , Colágeno Tipo III/análise , Colágeno Tipo III/metabolismo , Colágeno Tipo IV/análise , Colágeno Tipo IV/metabolismo , AMP Cíclico/metabolismo , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Ensaio de Imunoadsorção Enzimática , Homeostase/efeitos dos fármacos , Humanos , Ifosfamida/química , Ifosfamida/metabolismo , Isoquinolinas/farmacologia , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas/farmacologia
11.
J Vasc Res ; 49(3): 231-41, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22433677

RESUMO

BACKGROUND/AIMS: It is currently under debate whether aldosterone is able to induce fibrosis or whether it acts only as a cofactor under pathological conditions, e.g. as an elevated salt (NaCl) load. METHODS: We tested the interaction of 10 nM aldosterone, 15 mM NaCl and 1 µM ouabain using rat aorta smooth muscle cells (A10) with respect to the following parameters: necrosis, apoptosis, glucose-6-phosphate dehydrogenase (G6PD) and 6-phosphogluconate dehydrogenase activity, glutathione (GSH) content, collagen and fibronectin homeostasis and intracellular calcium distribution. RESULTS: Necrosis rates were increased after 48 h of incubation with aldosterone, salt or ouabain and in the combination of aldosterone and salt or ouabain. Apoptosis rates were decreased. A reduced defense capacity against oxidative stress was mirrored in the decreased G6PD activity and GSH content. Collagen III or fibronectin synthesis rates were unchanged, but gelatinase activity was increased resulting in a decreased media collagen III and fibronectin content. Calcium stores were increased by aldosterone in combination with ouabain. CONCLUSION: Aldosterone and salt per se can lead to cell injury that is aggravated in combination or with cardiotonic steroids. In cooperation with other vascular cells, this can generate a permissive milieu enabling aldosterone or salt to promote more extensive vascular injury.


Assuntos
Aldosterona/farmacologia , Aorta/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Ouabaína/farmacologia , Cloreto de Sódio/farmacologia , Animais , Aorta/citologia , Aorta/metabolismo , Cálcio/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Colágeno Tipo III/metabolismo , Fibronectinas/metabolismo , Glucosefosfato Desidrogenase/metabolismo , Glutationa/análise , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Ratos
12.
Biochim Biophys Acta ; 1803(5): 584-90, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20211660

RESUMO

The mineralocorticoid receptor (MR), a ligand-activated transcription factor expressed in various cell types (e.g. epithelial cells, neurons, smooth muscle cells, immune cells), plays important roles in neurohumoral, neuronal, cardiovascular, renal and intestinal function. Pathophysiological relevant signaling mechanisms include nongenomic pathways involving the EGF receptor (EGFR). We investigated whether a MR-EGFR colocalization may underlie the functional MR-EGFR interaction by coimmunoprecipitation, fluorescence resonance energy transfer (FRET) and confocal microscopy in a heterologous expression system. EGFR and a small fraction of MR colocalize at the cell membrane, independently of short time exposure (

Assuntos
Membrana Celular/metabolismo , Receptores ErbB/metabolismo , Rim/metabolismo , Mineralocorticoides/metabolismo , Receptores de Mineralocorticoides/metabolismo , Western Blotting , Células Cultivadas , Transferência Ressonante de Energia de Fluorescência , Imunofluorescência , Humanos , Imunoprecipitação , Rim/citologia , Ligação Proteica
13.
FASEB J ; 24(6): 2010-9, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20103717

RESUMO

We investigated the interaction of MR with cAMP-response element binding protein (CREB) and provide a mechanistic explanation and insights into the cellular relevance. MR --> CREB crosstalk was assessed in vascular smooth muscle cells and heterologous expression systems. Experiments were designed in a way that only one variable changed at a time and the respective vehicles served as controls. MR, but not GR, activation (aldosterone or hydrocortisone, IC(50), approximately 0.3 nM) inhibits CREB transcriptional activity induced by stimulation of beta1/2-adrenoceptors and adenylyl cyclase or addition of membrane-permeable cAMP up to 70% within 2 h after addition. The MR DNA-binding domain is not required for this inhibition. cAMP formation is virtually unchanged, whereas MR exerts a robust inhibition of CREB(S133) phosphorylation via calcineurin/PP2B activation without changes in PP2B-Aalpha or beta expression. In parallel, the PP2B-sensitive NFaT-pathway is activated. The inhibitory crosstalk attenuates CREB-induced glucose-6-phosphate dehydrogenase expression. Overall, transcriptional relevant MR --> CREB crosstalk occurs at the level of CREB phosphorylation by enhanced calcineurin activity, enables GRE-independent genomic signaling of MR, and is of potential pathophysiological relevance.


Assuntos
Aorta/metabolismo , Calcineurina/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Miócitos de Músculo Liso/metabolismo , Receptores de Mineralocorticoides/metabolismo , Aorta/citologia , Western Blotting , Calcineurina/genética , Células Cultivadas , AMP Cíclico/metabolismo , Modulador de Elemento de Resposta do AMP Cíclico/genética , Modulador de Elemento de Resposta do AMP Cíclico/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Ensaio de Imunoadsorção Enzimática , Glucosefosfato Desidrogenase/metabolismo , Humanos , Fosforilação , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Adrenérgicos/metabolismo , Receptores de Mineralocorticoides/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
14.
Am J Physiol Endocrinol Metab ; 292(6): E1790-800, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17311890

RESUMO

Aldosterone plays a key role in cardiovascular and renal injury. The underlying mechanisms are not completely understood. Because the epidermal growth factor receptor (EGFR) is involved in the development of fibrosis and vascular dysfunction, upregulation of EGFR expression by aldosterone-bound mineralocorticoid receptor (MR) is an attractive hypothesis. We investigated the effect of aldosterone on EGFR expression in the aorta of adrenalectomized rats and in human aorta smooth muscle cells (HAoSMC) in primary culture. Aldosterone, but not dexamethasone, stimulated EGFR expression in vivo in the aorta as well as in HAoSMC. EGFR degradation was not affected. Aldosterone-induced EGFR expression in HAoSMC was dose dependent and prevented by spironolactone. Furthermore, incubation of HAoSMC with aldosterone led to enhanced EGF-induced ERK1/2 phosphorylation and an EGFR-dependent increase in media fibronectin. EGFR promoter reporter gene assay as well as chromatin immunoprecipitation data indicate that MR interacts with the EGFR promoter. With deletion constructs we gained evidence that this interaction takes place between the hMR and the EGFR promoter regions 316-163 (stronger activation site, EC50 approximately 1.0 nM) and 163-1 (weaker activation site, EC50 approximately 0.7 nM), which do not comprise canonical glucocorticoid response elements and are not activated by the human glucocorticoid receptor. The interactions require in part the NH2-terminal domains of MR. ELISA-based transcription factor DNA binding assay with in vitro synthesized hMR suggest direct binding to region 163-1. Our results indicate that aldosterone leads to enhanced EGFR expression via an interaction with the EGFR promoter, which is MR specific and could contribute to the aldosterone-induced increase in fibronectin abundance.


Assuntos
Aldosterona/fisiologia , Aorta/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Regiões Promotoras Genéticas/fisiologia , Receptores de Mineralocorticoides/metabolismo , Adrenalectomia , Aldosterona/farmacologia , Animais , Células Cultivadas , Cromatina , DNA/metabolismo , Fragmentação do DNA , Fibronectinas/metabolismo , Humanos , Imunoprecipitação , Ligantes , Masculino , Estrutura Terciária de Proteína/fisiologia , Ratos , Ratos Wistar , Receptores de Mineralocorticoides/genética , Túnica Média/citologia , Túnica Média/metabolismo
15.
Pflugers Arch ; 454(3): 403-13, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17242955

RESUMO

The importance of aldosterone for cardiovascular diseases is well established. Most of the adverse effects seem to originate from its ability to produce vascular injury, including fibrosis. It is currently under debate whether aldosterone per se is able to induce fibrosis or whether it acts as a cofactor under pathological conditions. We tested whether aldosterone per se and in the presence of reactive oxygen stress (H(2)O(2)) enhances collagen abundance in human aortic smooth muscle cell (HAoSMC) media in primary culture and, if so, by which means. Collagen abundance, as well as epidermal growth factor receptor (EGFR) expression and ERK1/2 phosphorylation, was investigated by ELISA and Western blot. Collagenase activity and H(2)O(2) formation were determined by fluorometry and luminometry. Aldosterone alone did not affect collagen abundance but potentiated the stimulatory effect of low concentrations of H(2)O(2) (1-10 micromol/l). This effect disappeared when shedding of membrane-bound EGFR ligands was prevented by GM6001. EGFR expression and cellular EGF responsiveness were enhanced by aldosterone. Inhibition of the EGFR kinase (tyrphostin AG1478) prevented the increase of collagen. The increase in collagen abundance was prevented by blockade of the mineralocorticoid receptor (MR) and could be reproduced by MR transfection into Chinese hamster ovary cells. We conclude that aldosterone sensitizes HAoSMC for H(2)O(2)-induced increase of collagen abundance at least in part by enhanced EGFR expression.


Assuntos
Aldosterona/farmacologia , Colágeno/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Sequência de Bases , Células Cultivadas , Colagenases/metabolismo , Receptores ErbB/metabolismo , Gelatinases/metabolismo , Homeostase , Humanos , Peróxido de Hidrogênio/farmacologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores de Mineralocorticoides/genética
16.
Kidney Blood Press Res ; 29(5): 280-93, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17035713

RESUMO

Chloroacetaldehyde (CAA) is a metabolite of the alkylating agent ifosfamide (IFO) and putatively responsible for renal damage following anti-tumor therapy with IFO. Depletion of sulfhydryl (SH) groups has been reported from cell culture, animal and clinical studies. In this work the effect of CAA on human proximal tubule cells in primary culture (hRPTEC) was investigated. Toxicity of CAA was determined by protein content, cell number, LDH release, trypan blue exclusion assay and caspase-3 activity. Free thiols were measured by the method of Ellman. CAA reduced hRPTEC cell number and protein, induced a loss in free intracellular thiols and an increase in necrosis markers. CAA but not acrolein inhibited the cysteine proteases caspase-3, caspase-8 and cathepsin B. Caspase activation by cisplatin was inhibited by CAA. In cells stained with fluorescent dyes targeting lysosomes, CAA induced an increase in lysosomal size and lysosomal leakage. The effects of CAA on cysteine protease activities and thiols could be reproduced in cell lysate. Acidification, which slowed the reaction of CAA with thiol donors, could also attenuate effects of CAA on necrosis markers, thiol depletion and cysteine protease inhibition in living cells. Thus, CAA directly reacts with cellular protein and non-protein thiols, mediating its toxicity on hRPTEC. This effect can be reduced by acidification. Therefore, urinary acidification could be an option to prevent IFO nephropathy in patients.


Assuntos
Acetaldeído/análogos & derivados , Antineoplásicos Alquilantes/metabolismo , Antineoplásicos Alquilantes/toxicidade , Ifosfamida/metabolismo , Ifosfamida/toxicidade , Nefropatias/induzido quimicamente , Nefropatias/metabolismo , Compostos de Sulfidrila/toxicidade , Reagentes de Sulfidrila/metabolismo , Acetaldeído/química , Acetaldeído/metabolismo , Biomarcadores , Caspase 3/metabolismo , Caspase 8/metabolismo , Catepsina B/metabolismo , Contagem de Células , Morte Celular , Células Cultivadas , Corantes , Cisteína Endopeptidases/metabolismo , Humanos , Nefropatias/enzimologia , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/metabolismo , L-Lactato Desidrogenase/metabolismo , Microscopia de Fluorescência , Necrose , Compostos de Sulfidrila/química , Reagentes de Sulfidrila/química , Azul Tripano , gama-Glutamiltransferase/metabolismo
17.
Kidney Int ; 68(5): 2029-41, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16221203

RESUMO

BACKGROUND: Renal damage following chemotherapy with ifosfamide is attributed to the metabolic activation of the drug and the generation of chloroacetaldehyde (CAA). Little is known about the mechanism by which CAA impairs renal function. In this study the effect of CAA on intracellular Ca(2+) homeostasis in human renal proximal tubule cells (RPTEC) in primary culture was investigated. METHODS: Intracellular Ca(2+) was measured using the Ca(2+)-sensitive dye fura-2. Cell viability was determined by protein content and cell number. Oncotic and apoptotic cell death was assayed using trypan blue exclusion, caspase-3 activity, and 4',6-diamino-2-phenylindole (DAPI) staining. RESULTS: CAA (1.5 to 150 micromol/L) induced sustained elevations of intracellular free calcium ([Ca(2+)](i)) from 75 +/- 3 nmol/L to maximal 151 +/- 6 nmol/L. This effect was dependent on extracellular Ca(2+), but not Ca(2+) entry. The rise in [Ca(2+)](i) mediated by CAA could be attributed to inhibition of Na(+)-dependent extrusion of intracellular Ca(2+), indicating an inhibitory action of CAA on Na(+)/Ca(2+) exchange. Modulation of protein kinase A (PKA), but not protein kinase C (PKC) blunted the effect of CAA. Thus, CAA seems to inhibit Na(+)/Ca(2+) exchange by interaction with cyclic adenosine monophosphate (cAMP)-PKA-signaling. A 48-hour exposure to 15 micromol/L CAA significantly reduced cell number and protein content of RPTEC by induction of necrosis. This effect of 15 micromol/L CAA could be overcome by coadministration of the intracellular Ca(2+) chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid acetoxymethyl ester (BAPTA-AM). CONCLUSION: First, CAA inhibits the Na+/Ca2+-exchanger. Second, this effect is dependent on PKA. Third, CAA induces necrotic rather than apoptotic cell death. Finally, disturbed Ca(2+) homeostasis via Na(+)/Ca(2+) exchange contributes to the nephrotoxic action of CAA in RPTEC.


Assuntos
Acetaldeído/análogos & derivados , Antineoplásicos Alquilantes/toxicidade , Sinalização do Cálcio/efeitos dos fármacos , Ifosfamida/toxicidade , Nefropatias/induzido quimicamente , Túbulos Renais Proximais/efeitos dos fármacos , Acetaldeído/metabolismo , Cálcio/metabolismo , ATPases Transportadoras de Cálcio/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Linhagem Celular , Citosol/efeitos dos fármacos , Citosol/metabolismo , Espaço Extracelular/metabolismo , Homeostase/efeitos dos fármacos , Humanos , Concentração de Íons de Hidrogênio , Nefropatias/metabolismo , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/metabolismo , ATPases Transportadoras de Cálcio da Membrana Plasmática , RNA Mensageiro/análise , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático , Sódio/metabolismo , Trocador de Sódio e Cálcio/genética , Trocador de Sódio e Cálcio/metabolismo
18.
Am J Physiol Renal Physiol ; 286(6): F1226-31, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-14749256

RESUMO

Aldosterone enhances Na(+) reabsorption via epithelial Na(+) channels (ENaC). Aldosterone also stimulates the protein kinase ERK1/2- and the epidermal growth factor (EGF) receptor (EGFR)-signaling pathway. Yet EGF and ERK1/2 are known inhibitors of ENaC-mediated Na(+) reabsorption. In the present study, using the well-established Madin-Darby canine kidney C7 cell line, we tested the hypothesis that EGFR represents a negative-feedback control for chronic aldosterone-induced Na(+) reabsorption [amiloride-inhibitable short-circuit current (I(sc))]. Mineralocorticoid receptor expression was confirmed by RT-PCR and Western blot analysis. Aldosterone enhanced ERK1/2 phosphorylation in an EGFR-dependent way. Furthermore, aldosterone stimulated EGFR expression. Aldosterone (10 nmol/l) induced a small transient increase in I(sc) under control conditions. Inhibition of ERK1/2 phosphorylation with U-0126 (10 micromol/l) stimulated I(sc), indicating constitutive ENaC inhibition. Aldosterone exerted a significantly larger effect in the presence of U-0126 than without U-0126. EGF (10 microg/l) inhibited I(sc), whereas inhibition of EGFR kinase by tyrphostin AG-1478 (100 nmol/l) enhanced I(sc). Aldosterone was more effective in the presence of AG-1478 than without AG-1478. In summary, we propose that the EGFR-signaling cascade can serve as a negative-feedback control to limit the effect of aldosterone-induced Na(+) reabsorption.


Assuntos
Aldosterona/farmacologia , Receptores ErbB/fisiologia , Sódio/metabolismo , Animais , Western Blotting , Células Cultivadas , Cães , Ensaio de Imunoadsorção Enzimática , Canais Epiteliais de Sódio , Retroalimentação/fisiologia , Rim/citologia , Rim/efeitos dos fármacos , Rim/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Canais de Sódio/metabolismo , Canais de Sódio/fisiologia
19.
J Biol Chem ; 278(44): 43060-6, 2003 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-12939263

RESUMO

The steroid hormone aldosterone plays an important role during pathological tissue modifications, similar to cardiovascular or renal fibrosis. The underlying mechanisms for the pathological actions are not understood. Interaction of aldosterone with the epidermal growth factor (EGF) receptor is an attractive hypothesis to explain pathological tissue remodeling elicited by aldosterone, because (i) mineralocorticoids can sensitize cells for EGF, (ii) mineralocorticoid receptor (MR)-antagonists reduce EGFR-mRNA expression, (iii) EGFR itself supports the development of cardiovascular or renal fibrosis, and (iv) signaling elements involved in the pathological action of aldosterone (similar to ERK1/2 or NFkB) are typical downstream modules during EGF signaling. In addition, an interaction of aldosterone and EGF with respect to ERK1/2 activation has been described. Here we show that aldosterone stimulates EGFR expression in renal tissue of adrenalectomized rats and in human renal primary cell cultures. Furthermore, Chinese hamster ovary (CHO) cells normally devoid of EGFR or MR express EGFR after transfection with human MR (CHO-MR cells) but not after transfection with human glucocorticoid receptor (CHO-GR cells). In CHO-MR cells, EGFR-expression is up-regulated by aldosterone and inhibited by spironolactone. CHO-MR cells but not CHO-GR cells respond with ERK1/2 phosphorylation to EGF exposure. The responsiveness to other peptide hormones was virtually not affected. These data suggest that EGFR is an aldosterone-induced protein and is involved in the manifold (patho)biological actions of aldosterone.


Assuntos
Aldosterona/fisiologia , Receptores ErbB/biossíntese , Aldosterona/metabolismo , Animais , Sequência de Bases , Western Blotting , Células CHO , Células Cultivadas , Clonagem Molecular , Cricetinae , Ativação Enzimática , Ensaio de Imunoadsorção Enzimática , Humanos , Rim/citologia , Rim/metabolismo , Masculino , Microscopia de Fluorescência , Mineralocorticoides/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Dados de Sequência Molecular , Fosforilação , Regiões Promotoras Genéticas , Ratos , Ratos Wistar , Receptores de Glucocorticoides/metabolismo , Receptores de Mineralocorticoides/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência do Ácido Nucleico , Fatores de Tempo , Transfecção , Regulação para Cima
20.
J Biol Chem ; 277(48): 45892-7, 2002 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-12244120

RESUMO

The epidermal growth factor (EGF) regulates cell proliferation, differentiation, and ion transport using ERK1/2 as a downstream effector. Furthermore, the EGF receptor (EGFR) is involved in signaling by G-protein-coupled receptors, growth hormone, and cytokines via transactivation. It has been suggested that steroids interact with peptide hormones. Previously, we have shown that aldosterone modulates EGF responses in Madin-Darby canine kidney cells (Gekle, M., Freudinger, R., Mildenberger, S., and Silbernagl, S. (2002) Am. J. Physiol. 282, F669-F679). Here, we tested the hypothesis that human EGFR-1 can confer alternative aldosterone responsiveness with respect to ERK1/2 phosphorylation to Chinese hamster ovary cells, which do not express EGFR. Wild-type Chinese hamster ovary cells did not respond to EGF or aldosterone. After transfection of human EGFR-1, the cells responded to EGF, but not to aldosterone. However, when submaximal concentrations of EGF were used, nanomolar concentrations of aldosterone potentiated the action of EGF within minutes, resulting in a leftward shift of the EGF dose-response curve. This was not the case in mock-transfected cells. The EGFR kinase inhibitor tyrphostin AG1478 or the MEK1/2 inhibitor U0126 completely prevented the effect. Furthermore, aldosterone enhanced Tyr phosphorylation of c-Src and EGFR, and an inhibitor of cytosolic tyrosine kinases (4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d]pyriociaine) prevented the action of aldosterone. Our data show that aldosterone uses the EGF-EGFR-MEK1/2-ERK1/2 signaling cascade to elicit its alternative effects. In the presence of EGF, aldosterone leads to EGFR transactivation via cytosolic tyrosine kinases of the Src family.


Assuntos
Aldosterona/metabolismo , Receptores ErbB/fisiologia , Transdução de Sinais/fisiologia , Animais , Células CHO , Cricetinae , Cães , Ensaio de Imunoadsorção Enzimática , Receptores ErbB/genética , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA