20 research outputs found

    Expression and regulation of type 2A protein phosphatases and alpha4 signalling in cardiac health and hypertrophy

    Get PDF
    Abstract Cardiac physiology and hypertrophy are regulated by the phosphorylation status of many proteins, which is partly controlled by a poorly defined type 2A protein phosphatase-alpha4 intracellular signalling axis. Quantitative PCR analysis revealed that mRNA levels of the type 2A catalytic subunits were differentially expressed in H9c2 cardiomyocytes (PP2ACb[PP2ACa[PP4C[PP6C), NRVM (PP2ACb[PP2ACa = PP4C = PP6C), and adult rat ventricular myocytes (PP2ACa[ PP2ACb[PP6C[PP4C). Western analysis confirmed that all type 2A catalytic subunits were expressed in H9c2 cardiomyocytes; however, PP4C protein was absent in adult myocytes and only detectable following 26S proteasome inhibition. Short-term knockdown of alpha4 protein expression attenuated expression of all type 2A catalytic subunits. Pressure overload-induced left ventricular (LV) hypertrophy was associated with an increase in both PP2AC and alpha4 protein expression. Although PP6C expression was unchanged, expression of PP6C regulatory subunits (1) Sit4-associated protein 1 (SAP1) and (2) ankyrin repeat domain (ANKRD) 28 and 44 proteins was elevated, whereas SAP2 expression was reduced in hypertrophied LV tissue. Co-immunoprecipitation studies demonstrated that the interaction between alpha4 and PP2AC or PP6C subunits was either unchanged or reduced in hypertrophied LV tissue, respectively. Phosphorylation status of phospholemman (Ser63 and Ser68) was significantly increased by knockdown of PP2ACa, PP2ACb, or PP4C protein expression. DNA damage assessed by histone H2A.X phosphorylation (cH2A.X) in hypertrophied tissue remained unchanged. However, exposure of cardiomyocytes to H2O2 increased levels of cH2A.X which was unaffected by knockdown of PP6C expression, but was abolished by the short-term knockdown of alpha4 expression. This study illustrates the significance and altered activity of the type 2A protein phosphatase-alpha4 complex in healthy and hypertrophied myocardium

    Phosphorylation and Activation of the Plasma Membrane Na+/H+ Exchanger (NHE1) during Osmotic Cell Shrinkage

    Get PDF
    The Na+/H+ Exchanger isoform 1 (NHE1) is a highly versatile, broadly distributed and precisely controlled transport protein that mediates volume and pH regulation in most cell types. NHE1 phosphorylation contributes to Na+/H+ exchange activity in response to phorbol esters, growth factors or protein phosphatase inhibitors, but has not been observed during activation by osmotic cell shrinkage (OCS). We examined the role of NHE1 phosphorylation during activation by OCS, using an ideal model system, the Amphiuma tridactylum red blood cell (atRBC). Na+/H+ exchange in atRBCs is mediated by an NHE1 homolog (atNHE1) that is 79% identical to human NHE1 at the amino acid level. NHE1 activity in atRBCs is exceptionally robust in that transport activity can increase more than 2 orders of magnitude from rest to full activation. Michaelis-Menten transport kinetics indicates that either OCS or treatment with the phosphatase inhibitor calyculin-A (CLA) increase Na+ transport capacity without affecting transport affinity (Km = 44 mM) in atRBCs. CLA and OCS act non-additively to activate atNHE1, indicating convergent, phosphorylation-dependent signaling in atNHE1 activation. In situ 32P labeling and immunoprecipitation demonstrates that the net phosphorylation of atNHE1 is increased 4-fold during OCS coinciding with a more than 2-order increase in Na+ transport activity. This is the first reported evidence of increased NHE1 phosphorylation during OCS in any vertebrate cell type. Finally, liquid chromatography and mass spectrometry (LC-MS/MS) analysis of atNHE1 immunoprecipitated from atRBC membranes reveals 9 phosphorylated serine/threonine residues, suggesting that activation of atNHE1 involves multiple phosphorylation and/or dephosphorylation events

    Akt mediated mitochondrial protection in the heart: metabolic and survival pathways to the rescue

    Full text link

    Expression of type 2A protein phosphatases in cardiac health and disease

    No full text
    Cardiac physiology and hypertrophy is regulated by the phosphorylation status of many proteins, which is regulated (in part) by activity of the type 2A serine/threonine protein phosphatase family. Phosphatase activity of this family is conferred by the homologous PP2ACα/β, PP4C and PP6C catalytic subunits. Using quantitative PCR, gene expression of type 2A phosphatases showed that PP6C mRNA (5.07) expressed as relative units (RU) was higher than PP2ACα (3.52), PP2ACβ (3.24) and PP4C (4.54) in H9c2 cardiomyocytes. However, in adult rat ventricular myocytes (ARVM), PP4C mRNA (6.42) was higher than PP2ACα (2.52), PP2ACβ (3.93) and PP6C (4.96). Using Western immunoblotting, PP2ACα/β protein expression was similar in both H9c2 cardiomyocytes and ARVM. PP6C protein expression in ARVM was significantly higher (P<0.05) when compared to H9c2 cardiomyocytes, while PP4C protein expression in ARVM was undetectable. Additionally, we showed that 28 days of transverse aortic constriction in the mouse induced a ∼61% increase in the left ventricular (LV) mass index and a significant increase (p<0.01) in the expression of PP2ACα/β protein when compared to sham-operated LV myocardium. PP4C protein expression was not detectable in sham or hypertrophied LV myocardium, whereas PP6C protein expression was similar in both groups. This study illustrates the differences in the expression of the type 2A protein phosphatases in both H9c2 cardiomyocytes, ARVM and also highlights the importance of PP2A in cardiac pathological hypertrophy. Future work will aim to identify substrates of PP2ACα, PP2ACβ, PP4C and PP6C related to calcium regulation and hypertrophy in cardiomyocytes using small interfering RNA

    Regulation of PP2AC carboxylmethylation and cellular localisation by inhibitory Class G-Protein coupled receptors in cardiomyocytes

    Get PDF
    The enzymatic activity of the type 2A protein phosphatase (PP2A) holoenzyme, a major serine/threonine phosphatase in the heart, is conferred by its catalytic subunit (PP2AC). PP2AC activity and subcellular localisation can be regulated by reversible carboxylmethylation of its C-terminal leucine309 (leu309) residue. Previous studies have shown that the stimulation of adenosine type 1 receptors (A1.Rs) induces PP2AC carboxylmethylation and altered subcellular distribution in adult rat ventricular myocytes (ARVM). In the current study, we show that the enzymatic components that regulate the carboxylmethylation status of PP2AC, leucine carboxylmethyltransferase-1 (LCMT-1) and phosphatase methylesterase-1 (PME-1) are abundantly expressed in, and almost entirely localised in the cytoplasm of ARVM. The stimulation of Gi-coupled A1.Rs with N(6)-cyclopentyladenosine (CPA), and of other Gi-coupled receptors such as muscarinic M2 receptors (stimulated with carbachol) and angiotensin II AT2 receptors (stimulated with CGP42112) in ARVM, induced PP2AC carboxylmethylation at leu309 in a concentration-dependent manner. Exposure of ARVM to 10 µM CPA increased the cellular association between PP2AC and its methyltransferase LCMT-1, but not its esterase PME-1. Stimulation of A1.Rs with 10 µM CPA increased the phosphorylation of protein kinase B at ser473, which was abolished by the PI3K inhibitor LY294002 (20 µM), thereby confirming that PI3K activity is upregulated in response to A1.R stimulation by CPA in ARVM. A1.R-induced PP2AC translocation to the particulate fraction was abrogated by adenoviral expression of the alpha subunit (Gαt1) coupled to the transducin G-protein coupled receptor. A similar inhibitory effect on A1.R-induced PP2AC translocation was also seen with LY294002 (20 µM). These data suggest that in ARVM, A1.R-induced PP2AC translocation to the particulate fraction occurs through a GiPCR-Gβγ-PI3K mediated intracellular signalling pathway, which may involve elevated PP2AC carboxylmethylation at leu309

    Stabilised beta-catenin in postnatal ventricular myocardium leads to dilated cardiomyopathy and premature death

    Full text link
    Beta-catenin is a component of the intercalated disc in cardiomyocytes, but can also be involved in signalling and activation of gene transcription. We wanted to determine how long-term changes in beta-catenin expression levels would affect mature cardiomyocytes. Conditional transgenic mice that either lacked beta-catenin or that expressed a non-degradable form of beta-catenin in the adult ventricle were created. While mice lacking beta-catenin in the ventricle do not have an overt phenotype, mice expressing a non-degradable form develop dilated cardiomyopathy and do not survive beyond 5 months. A detailed analysis could reveal that this phenotype is correlated with a distinct localisation of beta-catenin in adult cardiomyocytes, which cannot be detected in the nucleus, no matter how much protein is present. Our report is the first study that addresses long-term effects of either the absence of beta-catenin or its stabilisation on ventricular cardiomyocytes and it suggests that beta-catenin's role in the nucleus may be of little significance in the healthy adult heart

    Inhibition of the Na-H + exchanger isoform-1 and the extracellular signal-regulated kinase induces apoptosis: A time course of events

    No full text
    Aims: The present study attempts to shed light on the role and the relative position of the Na +/H + exchanger isoform 1 (NHE1) and the extracellular signal-regulated kinase (ERK) in HEp-2 cell signaling pathways concerning a diverse range of cellular functions such as regulation of intracellular pH (pHi), DNA synthesis, production of reactive oxygen species (ROS) and apoptosis. Methods: Pharmacological inhibition with cariporide (highly specific inhibitor of NHE1) and PD98059 (specific inhibitor of the upstream activator of ERK) was implemented. Fluorescence spectrometry, atomic absorption spectrometry and ELISA methods were used in order to obtain the results. Results: NHE1 and ERK take part in all of the aforementioned cellular functions, as their inhibition had an effect on all of them. Additionally, inhibition of NHE1 resulted in ERK inhibition as well. Moreover, continuous inhibition of NHE1 or ERK for up to 24h led HEp-2 cells to apoptosis, as assessed through caspase-3 activation, DNA fragmentation and annexin-V binding levels. Conclusion: Our data shows a time course of events in relation to NHE1 and ERK and suggests the existence of a positive feedback loop between NHE1 and ERK which could pose a barrier against apoptosis. Copyright © 2006 S. Karger AG
    corecore