Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Circ Res ; 129(8): 804-820, 2021 10.
Article in English | MEDLINE | ID: mdl-34433292
2.
Proc Natl Acad Sci U S A ; 116(44): 22282-22287, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31619570

ABSTRACT

Sympathetic activation of ß-adrenoreceptors (ß-AR) represents a hallmark in the development of heart failure (HF). However, little is known about the underlying mechanisms of gene regulation. In human ventricular myocardium from patients with end-stage HF, we found high levels of phosphorylated histone 3 at serine-28 (H3S28p). H3S28p was increased by inhibition of the catecholamine-sensitive protein phosphatase 1 and decreased by ß-blocker pretreatment. By a series of in vitro and in vivo experiments, we show that the ß-AR downstream protein kinase CaM kinase II (CaMKII) directly binds and phosphorylates H3S28. Whereas, in CaMKII-deficient myocytes, acute catecholaminergic stimulation resulted in some degree of H3S28p, sustained catecholaminergic stimulation almost entirely failed to induce H3S28p. Genome-wide analysis of CaMKII-mediated H3S28p in response to chronic ß-AR stress by chromatin immunoprecipitation followed by massive genomic sequencing led to the identification of CaMKII-dependent H3S28p target genes. Forty percent of differentially H3S28p-enriched genomic regions were associated with differential, mostly increased expression of the nearest genes, pointing to CaMKII-dependent H3S28p as an activating histone mark. Remarkably, the adult hemoglobin genes showed an H3S28p enrichment close to their transcriptional start or end sites, which was associated with increased messenger RNA and protein expression. In summary, we demonstrate that chronic ß-AR activation leads to CaMKII-mediated H3S28p in cardiomyocytes. Thus, H3S28p-dependent changes may play an unexpected role for cardiac hemoglobin regulation in the context of sympathetic activation. These data also imply that CaMKII may be a yet unrecognized stress-responsive regulator of hematopoesis.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Heart Failure/metabolism , Hemoglobins/genetics , Histone Code , Histones/metabolism , Myocardium/metabolism , Sympathetic Nervous System/physiology , Adrenergic beta-Antagonists/pharmacology , Adult , Animals , Catecholamines/pharmacology , Cells, Cultured , Female , Heart Failure/genetics , Hemoglobins/metabolism , Humans , Male , Mice , Middle Aged , Phosphorylation , Rats , Sympathetic Nervous System/drug effects
3.
Sci Rep ; 8(1): 17711, 2018 12 07.
Article in English | MEDLINE | ID: mdl-30531830

ABSTRACT

Heart failure is the most common cause of morbidity and hospitalization in the western civilization. Protein phosphatases play a key role in the basal cardiac contractility and in the responses to ß-adrenergic stimulation with type-1 phosphatase (PP-1) being major contributor. We propose here that formation of transient disulfide bridges in PP-1α might play a leading role in oxidative stress response. First, we established an optimized workflow, the so-called "cross-over-read" search method, for the identification of disulfide-linked species using permutated databases. By applying this method, we demonstrate the formation of unexpected transient disulfides in PP-1α to shelter against over-oxidation. This protection mechanism strongly depends on the fast response in the presence of reduced glutathione. Our work points out that the dimerization of PP-1α involving Cys39 and Cys127 is presumably important for the protection of PP-1α active surface in the absence of a substrate. We finally give insight into the electron transport from the PP-1α catalytic core to the surface. Our data suggest that the formation of transient disulfides might be a general mechanism of proteins to escape from irreversible cysteine oxidation and to prevent their complete inactivation.


Subject(s)
Disulfides/metabolism , Glutathione/metabolism , Oxidative Stress/physiology , Phosphoric Monoester Hydrolases/metabolism , Animals , Catalytic Domain/physiology , Cysteine/metabolism , Dimerization , Electron Transport/physiology , Myocytes, Cardiac/metabolism , Oxidation-Reduction , Rats
5.
Basic Res Cardiol ; 112(4): 43, 2017 07.
Article in English | MEDLINE | ID: mdl-28597249

ABSTRACT

Protein phosphatase 1 (PP1) is a key regulator of important cardiac signaling pathways. Dysregulation of PP1 has been heavily implicated in cardiac dysfunctions. Accordingly, pharmacological targeting of PP1 activity is considered for therapeutic intervention in human cardiomyopathies. Recent evidence from animal models implicated previously unrecognized, isoform-specific activities of PP1 in the healthy and diseased heart. Therefore, this study examined the expression of the distinct PP1 isoforms PP1α, ß, and γ in human heart failure (HF) and atrial fibrillation (AF) and addressed the consequences of ß-adrenoceptor blocker (beta-blocker) therapy for HF patients with reduced ejection fraction on PP1 isoform expression. Using western blot analysis, we found greater abundance of PP1 isoforms α and γ but unaltered PP1ß levels in left ventricular myocardial tissues from HF patients as compared to non-failing controls. However, expression of all three PP1 isoforms was higher in atrial appendages from patients with AF compared to patients with sinus rhythm. Moreover, we found that in human failing ventricles, beta-blocker therapy was associated with lower PP1α abundance and activity, as indicated by higher phosphorylation of the PP1α-specific substrate eIF2α. Greater eIF2α phosphorylation is a known repressor of protein translation, and accordingly, we found lower levels of the endoplasmic reticulum (ER) stress marker Grp78 in the very same samples. We propose that isoform-specific targeting of PP1α activity may be a novel and innovative therapeutic strategy for the treatment of human cardiac diseases by reducing ER stress conditions.


Subject(s)
Atrial Fibrillation/enzymology , Heart Failure/enzymology , Myocardium/enzymology , Protein Phosphatase 1/metabolism , Adrenergic beta-Antagonists/therapeutic use , Adult , Aged , Aged, 80 and over , Atrial Fibrillation/pathology , Atrial Fibrillation/physiopathology , Case-Control Studies , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Eukaryotic Initiation Factor-2/metabolism , Female , HEK293 Cells , Heart Atria/enzymology , Heart Atria/pathology , Heart Failure/drug therapy , Heart Failure/pathology , Heart Failure/physiopathology , Heart Rate , Heart Ventricles/enzymology , Heart Ventricles/pathology , Heat-Shock Proteins/metabolism , Humans , Isoenzymes , Male , Middle Aged , Myocardium/pathology , Phosphorylation , Protein Phosphatase 1/genetics , Stroke Volume , Substrate Specificity , Transfection , Ventricular Function, Left
6.
Circ Heart Fail ; 10(5): e003840, 2017 May.
Article in English | MEDLINE | ID: mdl-28487342

ABSTRACT

BACKGROUND: Considerable evidence suggests that calcium/calmodulin-dependent protein kinase II (CaMKII) overactivity plays a crucial role in the pathophysiology of heart failure (HF), a condition characterized by excessive ß-adrenoceptor (ß-AR) stimulation. Recent studies indicate a significant cross talk between ß-AR signaling and CaMKII activation presenting CaMKII as a possible downstream mediator of detrimental ß-AR signaling in HF. In this study, we investigated the effect of chronic ß-AR blocker treatment on CaMKII activity in human and experimental HF. METHODS AND RESULTS: Immunoblot analysis of myocardium from end-stage HF patients (n=12) and non-HF subjects undergoing cardiac surgery (n=12) treated with ß-AR blockers revealed no difference in CaMKII activity when compared with non-ß-AR blocker-treated patients. CaMKII activity was judged by analysis of CaMKII expression, autophosphorylation, and oxidation and by investigating the phosphorylation status of CaMKII downstream targets. To further evaluate these findings, CaMKIIδC transgenic mice were treated with the ß1-AR blocker metoprolol (270 mg/kg*d). Metoprolol significantly reduced transgene-associated mortality (n≥29; P<0.001), attenuated the development of cardiac hypertrophy (-14±6% heart weight/tibia length; P<0.05), and strongly reduced ventricular arrhythmias (-70±22% premature ventricular contractions; P<0.05). On a molecular level, metoprolol expectedly decreased protein kinase A-dependent phospholamban and ryanodine receptor 2 phosphorylation (-42±9% for P-phospholamban-S16 and -22±7% for P-ryanodine receptor 2-S2808; P<0.05). However, this was paralled neither by a reduction in CaMKII autophosphorylation, oxidation, and substrate binding nor a change in the phosphorylation of CaMKII downstream target proteins (n≥11). The lack of CaMKII modulation by ß-AR blocker treatment was confirmed in healthy wild-type mice receiving metoprolol. CONCLUSIONS: Chronic ß-AR blocker therapy in patients and in a mouse model of CaMKII-induced HF is not associated with a change in CaMKII activity. Thus, our data suggest that the molecular effects of ß-AR blockers are not based on a modulation of CaMKII. Directly targeting CaMKII may, therefore, further improve HF therapy in addition to ß-AR blockade.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Heart Failure/metabolism , Metoprolol/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Disease Models, Animal , Echocardiography , Heart Failure/diagnosis , Heart Failure/drug therapy , Humans , Immunoblotting , Mice , Mice, Transgenic
7.
Naunyn Schmiedebergs Arch Pharmacol ; 390(8): 857-862, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28451724

ABSTRACT

Inhibitor-1 (I-1) modulates protein phosphatase 1 (PP1) activity and thereby counteracts the phosphorylation by kinases. I-1 is downregulated and deactivated in failing hearts, but whether its role is beneficial or detrimental remains controversial, and opposing therapeutic strategies have been proposed. Overactivity of Ca2+/calmodulin-dependent protein kinase II (CaMKII) with hyperphosphorylation of ryanodine receptors (RyR2) at the CaMKII-site is recognized to be central for heart failure and arrhythmias. Using an I-1-deficient mouse line as well as transfected cell lines, we investigated the effects of acute and chronic modulation of I-1 on CaMKII activity and RyR2 phosphorylation. We demonstrate that I-1 acutely modulates CaMKII by regulating PP1 activity. However, while ablation of I-1 should thus limit CaMKII-activation, we unexpectedly found exaggerated CaMKII-activation under ß-adrenergic stress upon chronic loss of I-1 in knockout mice. We unraveled that this is due to chronic upregulation of the exchange protein activated by cAMP (EPAC) leading to augmented CaMKII activation, and using computational modeling validated that an increase in EPAC expression can indeed explain our experimental findings. Interestingly, at the level of RyR2, the increase in PP1 activity more than outweighed the increase in CaMKII activity, resulting in reduced RyR phosphorylation at Ser-2814. Exaggerated CaMKII activation due to counterregulatory mechanisms upon loss of I-1 is an important caveat with respect to suggested therapeutic I-1-inhibition, as CaMKII overactivity has been heavily implicated in several cardiac pathologies.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Myocardium/metabolism , Proteins/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Adrenergic beta-1 Receptor Agonists , Animals , Dobutamine , Dogs , Echocardiography, Stress , Guanine Nucleotide Exchange Factors/metabolism , Mice, Knockout , Myocytes, Cardiac/metabolism , Phosphorylation , Protein Phosphatase 1/metabolism , Rats , Stress, Physiological
8.
J Mol Cell Cardiol ; 101: 116-126, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27639308

ABSTRACT

Phosphoproteomic studies have shown that about one third of all cardiac proteins are reversibly phosphorylated, affecting virtually every cellular signaling pathway. The reversibility of this process is orchestrated by the opposing enzymatic activity of kinases and phosphatases. Conversely, imbalances in subcellular protein phosphorylation patterns are a hallmark of many cardiovascular diseases including heart failure and cardiac arrhythmias. While numerous studies have revealed excessive beta-adrenergic signaling followed by deregulated kinase expression or activity as a major driver of the latter cardiac pathologies, far less is known about the beta-adrenergic regulation of their phosphatase counterparts. In fact, most of the limited knowledge stems from the detailed analysis of the endogenous inhibitor of the protein phosphatase 1 (I-1) in cellular and animal models. I-1 acts as a nodal point between adrenergic and putatively non-adrenergic cardiac signaling pathways and is able to influence widespread cellular functions of protein phosphatase 1 which are contributing to cardiac health and disease, e.g. Ca2+ handling, sarcomere contractility and glucose metabolism. Finally, nearly all of these studies agree that I-1 is a promising drug target on the one hand but the outcome of its pharmacological regulation maybe extremely context-dependent on the other hand, thus warranting for careful interpretation of past and future experimental results. In this respect we will: 1) comprehensively review the current knowledge about structural, functional and regulatory properties of I-1 within the heart 2) highlight current working hypothesis and potential I-1 mediated disease mechanisms 3) discuss state-of-the-art knowledge and future prospects of a potential therapeutic strategy targeting I-1 by restoring the balance of cardiac protein phosphorylation.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Myocytes, Cardiac/metabolism , Receptors, Adrenergic, beta/metabolism , Signal Transduction , Animals , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/metabolism , Heart Failure/drug therapy , Heart Failure/etiology , Heart Failure/metabolism , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Molecular Targeted Therapy , Organ Specificity
9.
Circ Res ; 119(9): 1004-1016, 2016 Oct 14.
Article in English | MEDLINE | ID: mdl-27553648

ABSTRACT

RATIONALE: Changes in redox potentials of cardiac myocytes are linked to several cardiovascular diseases. Redox alterations are currently mostly described qualitatively using chemical sensors, which however do not allow quantifying redox potentials, lack specificity, and the possibility to analyze subcellular domains. Recent advances to quantitatively describe defined redox changes include the application of genetically encoded redox biosensors. OBJECTIVE: Establishment of mouse models, which allow the quantification of the glutathione redox potential (EGSH) in the cytoplasm and the mitochondrial matrix of isolated cardiac myocytes and in Langendorff-perfused hearts based on the use of the redox-sensitive green fluorescent protein 2, coupled to the glutaredoxin 1 (Grx1-roGFP2). METHODS AND RESULTS: We generated transgenic mice with cardiac myocyte-restricted expression of Grx1-roGFP2 targeted either to the mitochondrial matrix or to the cytoplasm. The response of the roGFP2 toward H2O2, diamide, and dithiothreitol was titrated and used to determine the EGSH in isolated cardiac myocytes and in Langendorff-perfused hearts. Distinct EGSH were observed in the cytoplasm and the mitochondrial matrix. Stimulation of the cardiac myocytes with isoprenaline, angiotensin II, or exposure to hypoxia/reoxygenation additionally underscored that these compartments responded independently. A compartment-specific response was also observed 3 to 14 days after myocardial infarction. CONCLUSIONS: We introduce redox biosensor mice as a new tool, which allows quantification of defined alterations of EGSH in the cytoplasm and the mitochondrial matrix in cardiac myocytes and can be exploited to answer questions in basic and translational cardiovascular research.


Subject(s)
Biosensing Techniques/methods , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Myocytes, Cardiac/metabolism , Animals , Cells, Cultured , Heart/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Culture Techniques , Oxidation-Reduction , Oxygen Consumption/physiology
10.
Front Pharmacol ; 6: 270, 2015.
Article in English | MEDLINE | ID: mdl-26617522

ABSTRACT

Decades of cardiovascular research have shown that variable and flexible levels of protein phosphorylation are necessary to maintain cardiac function. A delicate balance between phosphorylated and dephosphorylated states of proteins is guaranteed by a complex interplay of protein kinases (PKs) and phosphatases. Serine/threonine phosphatases, in particular members of the protein phosphatase (PP) family govern dephosphorylation of the majority of these cardiac proteins. Recent findings have however shown that PPs do not only dephosphorylate previously phosphorylated proteins as a passive control mechanism but are capable to actively control PK activity via different direct and indirect signaling pathways. These control mechanisms can take place on (epi-)genetic, (post-)transcriptional, and (post-)translational levels. In addition PPs themselves are targets of a plethora of proteinaceous interaction partner regulating their endogenous activity, thus adding another level of complexity and feedback control toward this system. Finally, novel approaches are underway to achieve spatiotemporal pharmacologic control of PPs which in turn can be used to fine-tune misleaded PK activity in heart disease. Taken together, this review comprehensively summarizes the major aspects of PP-mediated PK regulation and discusses the subsequent consequences of deregulated PP activity for cardiovascular diseases in depth.

SELECTION OF CITATIONS
SEARCH DETAIL
...