Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Cell Mol Life Sci ; 81(1): 272, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38900158

RESUMEN

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.


Asunto(s)
Núcleo Celular , Receptores ErbB , Receptor de Angiotensina Tipo 1 , Receptores ErbB/metabolismo , Humanos , Receptor de Angiotensina Tipo 1/metabolismo , Receptor de Angiotensina Tipo 1/genética , Núcleo Celular/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Fosforilación , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Membrana Celular/metabolismo , Angiotensina II/metabolismo , Angiotensina II/farmacología , Factor de Crecimiento Epidérmico/metabolismo , Células HEK293 , Unión Proteica , Factor de Respuesta Sérica/metabolismo , Factor de Respuesta Sérica/genética
2.
Cell Mol Life Sci ; 79(1): 57, 2021 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-34921637

RESUMEN

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.


Asunto(s)
Regulación de la Expresión Génica , Receptor de Angiotensina Tipo 1/metabolismo , Receptores ErbB/metabolismo , Células HEK293 , Humanos , Receptor Cross-Talk , Transducción de Señal , Transcriptoma
3.
Cell Mol Life Sci ; 77(5): 903-918, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31312877

RESUMEN

MicroRNAs (miRs) contribute to different aspects of cardiovascular pathology, among others cardiac hypertrophy and atrial fibrillation. The aim of our study was to evaluate the impact of miR-221/222 on cardiac electrical remodeling. Cardiac miR expression was analyzed in a mouse model with altered electrocardiography parameters and severe heart hypertrophy. Next generation sequencing revealed 14 differentially expressed miRs in hypertrophic hearts, with miR-221 and -222 being the strongest regulated miR-cluster. This increase was restricted to cardiomyocytes and not observed in cardiac fibroblasts. Additionally, we evaluated the change of miR-221/222 in vivo in two models of pharmacologically induced heart hypertrophy (angiotensin II, isoprenaline), thereby demonstrating a stimulus-induced increase in miR-221/222 in vivo by angiotensin II but not by isoprenaline. Whole transcriptome analysis by RNA-seq and qRT-PCR validation revealed an enriched number of downregulated mRNAs coding for proteins located in the T-tubule, which are also predicted targets for miR-221/222. Among those, mRNAs were the L-type Ca2+ channel subunits as well as potassium channel subunits. We confirmed that both miRs target the 3'-untranslated regions of Cacna1c and Kcnj5. Furthermore, enhanced expression of these miRs reduced L-type Ca2+ channel and Kcnj5 channel abundance and function, which was analyzed by whole-cell patch clamp recordings or Western blot and flux measurements, respectively. miR-221 and -222 contribute to the regulation of L-type Ca2+ channels as well as Kcnj5 channels and, therefore, potentially contribute to disturbed cardiac excitation generation and propagation. Future studies will have to evaluate the pathophysiological and clinical relevance of aberrant miR-221/222 expression for electrical remodeling.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , MicroARNs/genética , Canales de Potasio/metabolismo , Animales , Canales de Calcio Tipo L/genética , Cardiomegalia/genética , Cardiomegalia/patología , Línea Celular , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/genética , Ratones , Ratones Noqueados , Miocitos Cardíacos/citología , Técnicas de Placa-Clamp , Canales de Potasio/genética
4.
Diabetologia ; 63(10): 2218-2234, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32548701

RESUMEN

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.


Asunto(s)
Aorta/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Angiopatías Diabéticas/genética , Nefropatías Diabéticas/genética , Receptores ErbB/genética , Músculo Liso Vascular/metabolismo , Obesidad/metabolismo , Factor de Respuesta Sérica/metabolismo , Actinas/metabolismo , Animales , Aorta/fisiopatología , Línea Celular , Diabetes Mellitus Tipo 2/fisiopatología , Angiopatías Diabéticas/metabolismo , Angiopatías Diabéticas/fisiopatología , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/fisiopatología , Dieta Alta en Grasa , Células HEK293 , Humanos , Hiperglucemia/metabolismo , Hiperglucemia/fisiopatología , Ratones , Ratones Noqueados , Músculo Liso Vascular/fisiopatología , Miocitos del Músculo Liso , Obesidad/fisiopatología , Transducción de Señal , Remodelación Vascular , Quinasas Asociadas a rho/metabolismo
5.
Biochim Biophys Acta ; 1863(7 Pt A): 1519-33, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27012600

RESUMEN

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.


Asunto(s)
Movimiento Celular , Receptores ErbB/metabolismo , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Adenosina Trifosfato/farmacología , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular , Células Cultivadas , Endotelina-1/farmacología , Activación Enzimática , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/agonistas , Receptores ErbB/deficiencia , Receptores ErbB/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibrosis , Regulación de la Expresión Génica , Genotipo , Inflamación/genética , Inflamación/metabolismo , Ligandos , Ratones Noqueados , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Estrés Oxidativo , Fenotipo , Fenilefrina/farmacología , Cultivo Primario de Células , Transducción de Señal , Trombina/farmacología , Factores de Tiempo , Vasopresinas/farmacología
6.
Clin Sci (Lond) ; 130(1): 19-33, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26438881

RESUMEN

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.


Asunto(s)
Angiotensina II , Presión Sanguínea , Receptores ErbB/deficiencia , Hipertensión/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Factores de Edad , Animales , Presión Sanguínea/efectos de los fármacos , Cardiomegalia/inducido químicamente , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatología , Quimiocina CCL2/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Receptores ErbB/genética , Eliminación de Gen , Humanos , Hipertensión/inducido químicamente , Hipertensión/genética , Hipertensión/fisiopatología , Hipertensión/prevención & control , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/fisiopatología , Miocitos del Músculo Liso/efectos de los fármacos , Fosforilación , Inhibidor 1 de Activador Plasminogénico/metabolismo , Transducción de Señal , Factores de Tiempo , Remodelación Vascular , Vasoconstrictores/farmacología
7.
Cell Physiol Biochem ; 33(4): 1106-16, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24732969

RESUMEN

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.


Asunto(s)
Acetaldehído/análogos & derivados , Matriz Extracelular/metabolismo , Túbulos Renales Proximales/efectos de los fármacos , Acetaldehído/farmacología , Antineoplásicos Alquilantes/química , Antineoplásicos Alquilantes/metabolismo , Calcio/metabolismo , Células Cultivadas , Colágeno Tipo III/análisis , Colágeno Tipo III/metabolismo , Colágeno Tipo IV/análisis , Colágeno Tipo IV/metabolismo , AMP Cíclico/metabolismo , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Ensayo de Inmunoadsorción Enzimática , Homeostasis/efectos de los fármacos , Humanos , Ifosfamida/química , Ifosfamida/metabolismo , Isoquinolinas/farmacología , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacología
8.
FASEB J ; 26(6): 2327-37, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22345406

RESUMEN

Recently it was shown that the mineralocorticoid receptor (MR) may exert part of its transcriptional activity by mediation of calcineurin (PP2B). Here we investigated the mechanism of interaction of MR with calcineurin and provide a new MR signaling pathway with potential physiological and pathophysiological relevance. MR → calcineurin crosstalk was assessed in a heterologous expression system (human embryonic kidney cells), which provides the opportunity for detailed mechanistic investigation. SiRNA knockdown experiments show that activated MR, but not GR, reduces CREB- and enhances NFaT-mediated transcriptional activation via the catalytic calcineurin subunit PP2BAß but not via PP2BAα. Altered PP2BAß expression, elevated cytosolic Ca(2+), activation of mitogen-activated kinase [p38, extracellular signal-regulated kinase (ERK) 1/2], or protein kinase C do not seem to be involved, whereas inhibition of the chaperone heat-shock protein 90 (HSP90) abrogated the effect of MR. Coimmunoprecipitation indicates the existence of protein complexes harboring MR and PP2BAß independent of MR activation but dependent on HSP90. Activated MR alters the subcellular distribution of PP2BAß, enhancing its nuclear fraction, and reduces mRNA expression of the endogenous inhibitor CAIN (calcineurin inhibitor) but not of RCAN1 (regulator of calcineurin). Overall, transcriptional relevant MR → calcineurin crosstalk occurs via the catalytic subunit PP2BAß, enables glucocorticoid response element-independent genomic signaling of MR, and is of potential pathophysiological relevance. Mechanistically, the crosstalk results from HSP90-mediated cytosolic protein complex formation, altered subcellular distribution, and altered endogenous inhibitor expression.


Asunto(s)
Calcineurina/metabolismo , Receptores de Mineralocorticoides/fisiología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Técnicas de Silenciamiento del Gen , Células HEK293 , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Humanos , Factores de Transcripción NFATC/metabolismo , ARN Interferente Pequeño/farmacología , Receptores de Mineralocorticoides/efectos de los fármacos
9.
Biomedicines ; 11(8)2023 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-37626737

RESUMEN

(1) Background: Obesity is associated with hypertension because of endocrine dysregulation of the adrenergic and the renin-angiotensin-aldosterone systems. The epidermal growth factor receptor (EGFR) is an important signaling hub in the cardiovascular system. In this study, we investigate the role of smooth muscle cell (VSMC) and endothelial cell (EC) EGFRs for blood pressure homeostasis and acute vascular reactivity in vivo. (2) Methods: Mice with deletion of the EGFR in the respective cell type received either a high-fat (HFD) or standard-fat diet (SFD) for 18 weeks. Intravascular blood pressure was measured via a Millar catheter in anesthetized animals upon vehicle load, angiotensin II (AII) and phenylephrine (PE) stimulation. (3) Results: We confirmed that deletion of the EGFR in VSMCs leads to reduced blood pressure and a most probably compensatory heart rate increase. EC-EGFR and VSMC-EGFR had only a minor impact on volume-load-induced blood pressure changes in lean as well as in obese wild-type animals. Regarding vasoactive substances, EC-EGFR seems to have no importance for angiotensin II action and counteracting HFD-induced prolonged blood pressure increase upon PE stimulation. VSMC-EGFR supports the blood pressure response to adrenergic and angiotensin II stimulation in lean animals. The responsiveness to AII and alpha-adrenergic stimulation was similar in lean and obese animals despite the known enhanced activity of the RAAS and the sympathetic nervous system under a high-fat diet. (4) Conclusions: We demonstrate that EGFRs in VSMCs and to a lesser extent in ECs modulate short-term vascular reactivity to AII, catecholamines and volume load in lean and obese animals.

10.
iScience ; 26(11): 108286, 2023 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-38026216

RESUMEN

Vascular smooth muscle cells (VSMC) are critical for the vascular tone, but they can also drive the development of vascular diseases when they lose their contractile phenotype and de-differentiate. Previous studies showed that the epidermal growth factor receptor (EGFR) of VSMC is critical for vascular health, but most of the underlying mechanisms by which VSMC-EGFR controls vascular fate have remained unknown. We combined RNA-sequencing and bioinformatics analysis to characterize the effect of EGFR-activation on the transcriptome of human primary VSMC (from different female donors) and to identify potentially affected cellular processes. Our results indicate that the activation of human VSMC-EGFR is sufficient to trigger a phenotypical switch toward a proliferative and inflammatory phenotype. The extent of this effect is nonetheless partly donor-dependent. Our hypothesis-generating study thus provides a first insight into mechanisms that could partly explain variable susceptibilities to vascular diseases in between individuals.

11.
Sci Rep ; 13(1): 22827, 2023 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-38129563

RESUMEN

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.


Asunto(s)
Vasos Coronarios , Células Endoteliales , Humanos , Femenino , Vasos Coronarios/metabolismo , Células Endoteliales/metabolismo , Células Cultivadas , Aorta , Receptores ErbB/metabolismo , Inflamación/metabolismo , Endotelio Vascular/metabolismo
12.
J Vasc Res ; 49(3): 231-41, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22433677

RESUMEN

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.


Asunto(s)
Aldosterona/farmacología , Aorta/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Ouabaína/farmacología , Cloruro de Sodio/farmacología , Animales , Aorta/citología , Aorta/metabolismo , Calcio/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Colágeno Tipo III/metabolismo , Fibronectinas/metabolismo , Glucosafosfato Deshidrogenasa/metabolismo , Glutatión/análisis , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Ratas
13.
Biochim Biophys Acta ; 1803(5): 584-90, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20211660

RESUMEN

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 (

Asunto(s)
Membrana Celular/metabolismo , Receptores ErbB/metabolismo , Riñón/metabolismo , Mineralocorticoides/metabolismo , Receptores de Mineralocorticoides/metabolismo , Western Blotting , Células Cultivadas , Transferencia Resonante de Energía de Fluorescencia , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Riñón/citología , Unión Proteica
14.
FASEB J ; 24(6): 2010-9, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20103717

RESUMEN

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.


Asunto(s)
Aorta/metabolismo , Calcineurina/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Miocitos del Músculo Liso/metabolismo , Receptores de Mineralocorticoides/metabolismo , Aorta/citología , Western Blotting , Calcineurina/genética , Células Cultivadas , AMP Cíclico/metabolismo , Modulador del Elemento de Respuesta al AMP Cíclico/genética , Modulador del Elemento de Respuesta al AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Ensayo de Inmunoadsorción Enzimática , Glucosafosfato Deshidrogenasa/metabolismo , Humanos , Fosforilación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Adrenérgicos/metabolismo , Receptores de Mineralocorticoides/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
15.
Biomedicines ; 9(9)2021 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-34572303

RESUMEN

Mineralocorticoids (e.g., aldosterone) support chronic inflammatory tissue damage, including glomerular mesangial injury leading to glomerulosclerosis. Furthermore, aldosterone leads to activation of the extracellular signal-regulated kinases (ERK1/2) in rat glomerular mesangial cells (GMC). Because ERK1/2 can affect cellular pH homeostasis via activation of Na+/H+-exchange (NHE) and the resulting cellular alkalinization may support proliferation, we tested the hypothesis that aldosterone affects pH homeostasis and thereby cell proliferation as well as collagen secretion also in primary rat GMC. Cytoplasmic pH and calcium were assessed by single-cell fluorescence ratio imaging, using the dyes BCECF or FURA2, respectively. Proliferation was determined by cell counting, thymidine incorporation and collagen secretion by collagenase-sensitive proline incorporation and ERK1/2-phosphorylation by Western blot. Nanomolar aldosterone induces a rapid cytosolic alkalinization which is prevented by NHE inhibition (10 µmol/L EIPA) and by blockade of the mineralocorticoid receptor (100 nmol/L spironolactone). pH changes were not affected by inhibition of HCO3- transporters and were not dependent on HCO3-. Aldosterone enhanced ERK1/2 phosphorylation and inhibition of ERK1/2-phosphorylation (10 µmol/L U0126) prevented aldosterone-induced alkalinization. Furthermore, aldosterone induced proliferation of GMC and collagen secretion, both of which were prevented by U0126 and EIPA. Cytosolic calcium was not involved in this aldosterone action. In conclusion, our data show that aldosterone can induce GMC proliferation via a MR and ERK1/2-mediated activation of NHE with subsequent cytosolic alkalinization. GMC proliferation leads to glomerular hypercellularity and dysfunction. This effect presents a possible mechanism contributing to mineralocorticoid receptor-induced pathogenesis of glomerular mesangial injury during chronic kidney disease.

16.
Sci Rep ; 11(1): 7269, 2021 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-33790318

RESUMEN

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.


Asunto(s)
Aorta Abdominal/metabolismo , Dieta Alta en Grasa/efectos adversos , Endotelio Vascular/metabolismo , Receptores ErbB/metabolismo , Riñón/metabolismo , Obesidad/metabolismo , Animales , Receptores ErbB/genética , Ratones , Ratones Noqueados , Obesidad/inducido químicamente , Obesidad/genética
17.
Br J Pharmacol ; 175(14): 2956-2967, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29682743

RESUMEN

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.


Asunto(s)
Eplerenona/uso terapéutico , Cirrosis Hepática/tratamiento farmacológico , Antagonistas de Receptores de Mineralocorticoides/uso terapéutico , Receptores de Mineralocorticoides/fisiología , Animales , Tetracloruro de Carbono , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Eplerenona/farmacología , Células HEK293 , Hepatocitos/metabolismo , Humanos , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Masculino , Antagonistas de Receptores de Mineralocorticoides/farmacología , ARN Mensajero/metabolismo , Ratas Wistar , Receptores de Mineralocorticoides/genética
18.
Mol Cell Endocrinol ; 264(1-2): 35-43, 2007 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-17113706

RESUMEN

The mineralocorticcoid receptor (MR) plays an important role in salt and water homeostasis as well as during cardiovascular and renal fibrosis but little is known regarding its modulation by other signaling pathways. To investigate a possible modulation under controlled conditions we used human embryonic kidney (HEK) cells (devoid of endogenous MR) transfected with the human MR and measured transactivation with a GRE-SEAP-reporter construct. MR was compared to the glucocorticoid receptor (GR) as well as to MR lacking the N-terminal domains AB (MR(CDEF)). Chelation of cytosolic Ca2+ enhanced MR activity and SGK1-expression, whereas elevation of cytosolic Ca2+ with ionomycin or thapsigargin reduced MR activity. GR activity was not affected by ionomycin or thapsigargin. MR(CDEF) activity was not affected by chelation or elevation of cytosolic Ca2+. Inhibition of ERK1/2 activation by U0126 or activation of PKA by cAMP, previously shown to modulate MR and GR activity, did not affect MR(CDEF) activity either. H2O2<500micromol/l did not affect basal nor hormone-induced reporter activity. Higher concentrations exerted the same relative inhibitory effect on GRE-SEAP-activity under basal conditions as in the presence of aldosterone-stimulated MR and elicited cytotoxic effects. Our data indicate that the genomic function of MR can be modulated by cytosolic Ca2+, PKA and ERK1/2 via an interaction with the AB-domain. H2O2 seems not to affect relative MR activity directly under our experimental conditions.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Peróxido de Hidrógeno/farmacología , Oxidantes/farmacología , Receptores de Mineralocorticoides/biosíntesis , Elementos de Respuesta/fisiología , Calcio/metabolismo , Señalización del Calcio/fisiología , Línea Celular , Quelantes/farmacología , Relación Dosis-Respuesta a Droga , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Humanos , Ionomicina/farmacología , Ionóforos/farmacología , Riñón/citología , Riñón/metabolismo , Proteínas Quinasas/metabolismo , Receptores de Glucocorticoides/biosíntesis
19.
Mol Endocrinol ; 19(7): 1697-710, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15761031

RESUMEN

The steroid hormone aldosterone is important for salt and water homeostasis as well as for pathological tissue modifications in the cardiovascular system and the kidney. The mechanisms of action include a classical genomic pathway, but physiological relevant nongenotropic effects have also been described. Unlike for estrogens or progesterone, the mechanisms for these nongenotropic effects are not well understood, although pharmacological studies suggest a role for the mineralocorticoid receptor (MR). Here we investigated whether the MR contributes to nongenotropic effects. After transfection with human MR, aldosterone induced a rapid and dose-dependent phosphorylation of ERK1/2 and c-Jun NH2-terminal kinase (JNK) 1/2 kinases in Chinese hamster ovary or human embryonic kidney cells, which was reduced by the MR-antagonist spironolactone and involved cSrc kinase as well as the epidermal growth factor receptor. In primary human aortic endothelial cells, similar results were obtained for ERK1/2 and JNK1/2. Inhibition of MAPK kinase (MEK) kinase but not of protein kinase C prevented the rapid action of aldosterone and also reduced aldosterone-induced transactivation, most probably due to impaired nuclear-cytoplasmic shuttling of MR. Cytosolic Ca2+ was increased by aldosterone in mock- and in human MR-transfected cells to the same extend due to Ca2+ influx, whereas dexamethasone had virtually no effect. Spironolactone did not prevent the Ca2+ response. We conclude that some nongenotropic effects of aldosterone are MR dependent and others are MR independent (e.g. Ca2+), indicating a higher degree of complexity of rapid aldosterone signaling. According to this model, we have to distinguish three aldosterone signaling pathways: 1) genomic via MR, 2) nongenotropic via MR, and 3) nongenotropic MR independent.


Asunto(s)
Aldosterona/fisiología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Receptores de Mineralocorticoides/metabolismo , Transducción de Señal , Aldosterona/farmacología , Animales , Células CHO , Calcio/metabolismo , Cricetinae , Cricetulus , Citosol/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/enzimología , Homeostasis , Humanos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Fosforilación , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Receptores de Mineralocorticoides/genética
20.
Mol Nutr Food Res ; 49(1): 31-7, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15580662

RESUMEN

Ochratoxin A (OTA) is a nephrotoxic mycotoxin. There is evidence that OTA leads to cortical interstitial nephropathies in humans, associated with fibrosis. No data are available on the effect of OTA-induced collagen secretion from renal cortical cells. As kidney cortex mainly consists of proximal tubules, we investigated the effect of OTA on particular collagens (I, III, IV) in a well-established proximal tubular cell line (opossum kidney (OK) cells) and in primary cultured human renal proximal tubular epithelial cells (RPTECs). In fibroblasts, OTA neither exerted toxic effects nor induced collagen secretion, most probably due to the absence of suitable uptake mechanisms. OTA exerted time- and dose-dependent toxicity in both OK cells and human RPTECs. Moreover, OTA induced collagen secretion in a time- and dose-dependent manner in both cell types. In opposite to transforming growth factor beta1 (TGF-beta1), OTA incubation induced increased apical secretion of the basement membrane collagen IV. This might be evidence for a loss of cellular polarity after OTA incubation. We conclude that in proximal tubular cells, OTA is able to induce extracellular matrix deposition. As collagen secretion was also inducible in primary cultured human RPTECs, we hypothesize that OTA-induced extracellular matrix deposition by proximal tubular cells may be of importance in generation of renal diseases in humans which are under suspicion of being induced by OTA.


Asunto(s)
Colágeno/metabolismo , Túbulos Renales Proximales/metabolismo , Ocratoxinas/toxicidad , Animales , Línea Celular , Línea Celular Transformada , Colágeno Tipo I/metabolismo , Colágeno Tipo III/metabolismo , Colágeno Tipo IV/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Túbulos Renales Proximales/efectos de los fármacos , Ocratoxinas/administración & dosificación , Zarigüeyas , Factor de Crecimiento Transformador beta/farmacología , Factor de Crecimiento Transformador beta1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA