Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Acta Physiol (Oxf) ; 233(3): e13715, 2021 11.
Article in English | MEDLINE | ID: mdl-34228904

ABSTRACT

AIM: This study investigates the role of calcineurin for angiotensin II (AngII)-induced vascular remodelling with the help of a mouse model lacking the catalytic beta subunit of calcineurin (PPP3CB KO). METHODS: Wildtype (WT) and PPP3CB KO mice were treated for 4 weeks with AngII followed by assessment of blood pressure, histological evaluation of aortas and mRNA analysis of aortic genes PPP3CB-dependently regulated by AngII. Primary murine vascular smooth muscle cells (VSMCs) were used for qPCR, ELISA and Western Blot experiments as well as wound healing and cell proliferation assays. RESULTS: Upon AngII treatment, PPP3CB KO mice showed less aortic media thickening, lumen dilation and systolic blood pressure compared to WT mice. Next-generation sequencing data of aortic tissue indicated an increase in extracellular matrix components (EMCs), cell migration and cell proliferation. A PPP3CB-dependent increase in EMC was confirmed by qPCR in aorta and VSMCs. PPP3CB-dependent stimulation of VSMC migration could be verified by wound healing assays but markers of enhanced cell proliferation were only detectable in aortic tissue of WT mice but not in isolated WT or KO VSMCs. We could demonstrate in VSMCs with pharmacological inhibitors that PPP3CB leads to enhanced heparin-binding EGF-like growth factor (HB-EGF) secretion, epidermal growth factor receptor (EGFR) activation and consecutive stimulation of transforming growth factor ß(TGFß) and connective tissue growth factor (CTGF) signalling that enhances collagen expression. CONCLUSION: AngII-induced vascular remodelling involves PPP3CB, which leads to enhanced EMC production, VSMC migration and sustained increase in systolic blood pressure via HBEGF/EGFR-TGFß-CTGF signalling.


Subject(s)
Angiotensin II , Vascular Remodeling , Animals , Calcineurin , ErbB Receptors , Mice , Myocytes, Smooth Muscle
2.
Cell Mol Life Sci ; 77(5): 903-918, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31312877

ABSTRACT

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.


Subject(s)
Calcium Channels, L-Type/metabolism , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , MicroRNAs/genetics , Potassium Channels/metabolism , Animals , Calcium Channels, L-Type/genetics , Cardiomegaly/genetics , Cardiomegaly/pathology , Cell Line , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , Mice , Mice, Knockout , Myocytes, Cardiac/cytology , Patch-Clamp Techniques , Potassium Channels/genetics
3.
Sci Rep ; 7(1): 15340, 2017 11 10.
Article in English | MEDLINE | ID: mdl-29127314

ABSTRACT

The pathogenesis of cardiovascular diseases is a multifunctional process in which the mineralocorticoid receptor (MR), a ligand-dependent transcription factor, is involved as proven by numerous clinical studies. The development of pathophysiological MR actions depends on the existence of additional factors e.g. inflammatory cytokines and seems to involve posttranslational MR modifications e.g. phosphorylation. Casein kinase 2 (CK2) is a ubiquitously expressed multifunctional serine/threonine kinase that can be activated under inflammatory conditions as the MR. Sequence analysis and inhibitor experiments revealed that CK2 acts as a positive modulator of MR activity by facilitating MR-DNA interaction with subsequent rapid MR degradation. Peptide microarrays and site-directed mutagenesis experiments identified the highly conserved S459 as a functionally relevant CK2 phosphorylation site of the MR. Moreover, MR-CK2 protein-protein interaction mediated by HSP90 was shown by co-immunoprecipitation. During inflammation, cytokine stimulation led to a CK2-dependent increased expression of proinflammatory genes. The additional MR activation by aldosterone during cytokine stimulation augmented CK2-dependent NFκB signaling which enhanced the expression of proinflammatory genes further. Overall, in an inflammatory environment the bidirectional CK2-MR interaction aggravate the existing pathophysiological cellular situation.


Subject(s)
Aldosterone/pharmacology , Casein Kinase II/metabolism , Gene Expression Regulation/drug effects , Receptors, Mineralocorticoid/metabolism , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Casein Kinase II/genetics , HEK293 Cells , Humans , NF-kappa B/genetics , NF-kappa B/metabolism , Receptors, Mineralocorticoid/genetics
4.
Nucleic Acids Res ; 41(17): 8045-60, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23821666

ABSTRACT

The mineralocorticoid receptor (MR) is a ligand-induced transcription factor belonging to the steroid receptor family and involved in water-electrolyte homeostasis, blood pressure regulation, inflammation and fibrosis in the renocardiovascular system. The MR shares a common hormone-response-element with the glucocorticoid receptor but nevertheless elicits MR-specific effects including enhanced epidermal growth factor receptor (EGFR) expression via unknown mechanisms. The EGFR is a receptor tyrosine kinase that leads to activation of MAP kinases, but that can also function as a signal transducer for other signaling pathways. In the present study, we mechanistically investigate the interaction between a newly discovered MR- but not glucocorticoid receptor- responsive-element (=MRE1) of the EGFR promoter, specificity protein 1 (SP1) and MR to gain general insights into MR-specificity. Biological relevance of the interaction for EGFR expression and consequently for different signaling pathways in general is demonstrated in human, rat and murine vascular smooth muscle cells and cells of EGFR knockout mice. A genome-wide promoter search for identical binding regions followed by quantitative PCR validation suggests that the identified MR-SP1-MRE1 interaction might be applicable to other genes. Overall, a novel principle of MR-specific gene expression is explored that applies to the pathophysiologically relevant expression of the EGFR and potentially also to other genes.


Subject(s)
ErbB Receptors/genetics , Receptors, Mineralocorticoid/metabolism , Response Elements , Sp1 Transcription Factor/metabolism , Transcription, Genetic , Aldosterone/pharmacology , Animals , Base Sequence , Binding Sites , Cells, Cultured , ErbB Receptors/metabolism , HEK293 Cells , Humans , Mice , Mice, Knockout , Molecular Sequence Data , Promoter Regions, Genetic , Protein Structure, Tertiary , Rats , Receptors, Mineralocorticoid/chemistry , Signal Transduction , Sp1 Transcription Factor/antagonists & inhibitors , Sp3 Transcription Factor/metabolism , Up-Regulation
5.
Free Radic Biol Med ; 53(5): 1088-100, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22749806

ABSTRACT

The mineralocorticoid receptor (MR) plays an important role in salt and water homeostasis and pathological tissue modifications, such as cardiovascular and renal fibrosis. Importantly, MR activation by aldosterone per se is not sufficient for the deleterious effects but requires the additional presence of a certain pathological milieu. Phenomenologically, this milieu could be generated by enhanced nitrosative stress. However, little is known regarding the modulation of MR transcriptional activity in a pathological milieu. The glucocorticoid receptor (GR), the closest relative of the MR, binds to the same hormone-response element but elicits protective effects on the cardiovascular system. To investigate the possible modulation of MR and GR by nitrosative stress under controlled conditions we used human embryonic kidney (HEK) cells and measured MR and GR transactivation after stimulation with the nitric oxide (NO)-donor SNAP and the peroxynitrite-donor Sin-1. In the presence of corticosteroids NO led to a general reduced corticosteroid receptor activity by repression of corticosteroid receptor-DNA interaction. The NO-induced diminished transcriptional MR activity was most pronounced during stimulation with physiological aldosterone concentrations, suggesting that NO treatment prevented its pathophysiological overactivation. In contrast, single peroxynitrite administration specifically induced the MR transactivation activity whereas genomic GR activity remained unchanged. Mechanistically, peroxynitrite permitted nuclear MR translocation whereas the cytosolic GR distribution was unaffected. Consequently, peroxynitrite represents a MR-specific aldosterone mimetic. In summary, our data indicate that the genomic function of corticosteroid receptors can be modulated by nitrosative stress which may induce the shift from physiological toward pathophysiological MR effects.


Subject(s)
Molsidomine/analogs & derivatives , Oxidative Stress/drug effects , Receptors, Mineralocorticoid/metabolism , S-Nitroso-N-Acetylpenicillamine/pharmacology , Cells, Cultured , HEK293 Cells , Humans , Molsidomine/pharmacology , Nitric Oxide/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Receptors, Mineralocorticoid/genetics
6.
Chem Biol ; 19(6): 742-51, 2012 Jun 22.
Article in English | MEDLINE | ID: mdl-22726688

ABSTRACT

The mineralocorticoid receptor (MR), a member of the steroid receptor superfamily, regulates water-electrolyte balance and mediates pathophysiological effects in the renocardiovascular system. Previously, it was assumed that after binding aldosterone, the MR dissociates from HSP90, forms homodimers, and then translocates into the nucleus where it acts as a transcription factor (Guiochon-Mantel et al., 1989; Robertson et al., 1993; Savory et al., 2001). We found that, during aldosterone-induced nuclear translocation, MR is bound to HSP90 both in the cytosol and the nucleus. Homodimerization measured by eBRET and FRET takes place when the MR is already predominantly nuclear. In vitro binding of MR to DNA was independent of ligand but could be partially inhibited by geldanamycin. Overall, here we provide insights into classical MR signaling necessary for elucidating the mechanisms of pathophysiological MR effects and MR specificity.


Subject(s)
Cell Nucleus/metabolism , Protein Multimerization , Receptors, Mineralocorticoid/metabolism , Signal Transduction , Benzoquinones/pharmacology , Bioluminescence Resonance Energy Transfer Techniques , Cells, Cultured , DNA/metabolism , Fluorescence Resonance Energy Transfer , HEK293 Cells , HSP90 Heat-Shock Proteins/metabolism , Humans , Lactams, Macrocyclic/pharmacology , Mineralocorticoid Receptor Antagonists/pharmacology , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...