Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Ábel Perjés is active.

Publication


Featured researches published by Ábel Perjés.


PLOS ONE | 2014

Apelin Increases Cardiac Contractility via Protein Kinase Cε- and Extracellular Signal-Regulated Kinase-Dependent Mechanisms

Ábel Perjés; Réka Skoumal; Olli Tenhunen; Attila Kónyi; Mihály Simon; Iván G. Horváth; Risto Kerkelä; Heikki Ruskoaho; István Szokodi

Background Apelin, the endogenous ligand for the G protein-coupled apelin receptor, is an important regulator of the cardiovascular homoeostasis. We previously demonstrated that apelin is one of the most potent endogenous stimulators of cardiac contractility; however, its underlying signaling mechanisms remain largely elusive. In this study we characterized the contribution of protein kinase C (PKC), extracellular signal-regulated kinase 1/2 (ERK1/2) and myosin light chain kinase (MLCK) to the positive inotropic effect of apelin. Methods and Results In isolated perfused rat hearts, apelin increased contractility in association with activation of prosurvival kinases PKC and ERK1/2. Apelin induced a transient increase in the translocation of PKCε, but not PKCα, from the cytosol to the particulate fraction, and a sustained increase in the phosphorylation of ERK1/2 in the left ventricle. Suppression of ERK1/2 activation diminished the apelin-induced increase in contractility. Although pharmacological inhibition of PKC attenuated the inotropic response to apelin, it had no effect on ERK1/2 phosphorylation. Moreover, the apelin-induced positive inotropic effect was significantly decreased by inhibition of MLCK, a kinase that increases myofilament Ca2+ sensitivity. Conclusions Apelin increases cardiac contractility through parallel and independent activation of PKCε and ERK1/2 signaling in the adult rat heart. Additionally MLCK activation represents a downstream mechanism in apelin signaling. Our data suggest that, in addition to their role in cytoprotection, modest activation of PKCε and ERK1/2 signaling improve contractile function, therefore these pathways represent attractive possible targets in the treatment of heart failure.


Journal of Molecular and Cellular Cardiology | 2011

Role of reactive oxygen species in the regulation of cardiac contractility

Anna Maria Kubin; Réka Skoumal; Pasi Tavi; Attila Kónyi; Ábel Perjés; Hanna Leskinen; Heikki Ruskoaho; István Szokodi

Increased production of reactive oxygen species (ROS) has been linked to the pathogenesis of contractile dysfunction in heart failure. However, it is unclear whether ROS can regulate physiological cellular processes in the myocardium. Here, we characterized the role of endogenous ROS production in the acute regulation of cardiac contractility in the intact rat heart. In isolated perfused rat hearts, endothelin-1 (ET-1, 1nmol/L) stimulated ROS formation in the left ventricle, which was prevented by the antioxidant N-acetylcysteine and the NAD(P)H oxidase inhibitor apocynin. N-acetylcysteine, the superoxide dismutase mimetic MnTMPyP, and apocynin significantly attenuated ET-1-mediated inotropic effect, which was accompanied by inhibition of extracellular signal regulated kinase 1/2 (ERK1/2) phosphorylation. Moreover, the mitochondrial K(ATP) channel blocker 5-HD, and the mitochondrial large conductance calcium activated potassium channel blocker paxilline, but not the sarcolemmal K(ATP) channel blocker HMR 1098 attenuated the inotropic response to ET-1. However, ET-1-induced ROS generation was not abolished by inhibiting mitochondrial K(ATP) channel opening. In contrast to ET-1 stimulation, the positive inotropic effect of β(1)-adrenergic receptor agonist dobutamine (250nmol/L) was significantly augmented by N-acetylcysteine and apocynin. Moreover, dobutamine-induced phospholamban phosphorylation was markedly enhanced by apocynin. In conclusion, NAD(P)H oxidase-derived ROS play a physiological role in the acute regulation of cardiac contractility in the intact rat heart. Our results reveal that ET-1-induced increase in cardiac contractility is partially dependent on enhanced ROS generation, which in turn, activates the ERK1/2 pathway. On the other hand, β-adrenergic receptor-induced positive inotropic effect and phospholamban phosphorylation is enhanced by NAD(P)H oxidase inhibition.


Basic Research in Cardiology | 2016

Characterization of apela, a novel endogenous ligand of apelin receptor, in the adult heart

Ábel Perjés; Teemu Kilpiö; Johanna Ulvila; Johanna Magga; Tarja Alakoski; Zoltan Szabo; Laura Vainio; Eveliina Halmetoja; Olli Vuolteenaho; Ulla E. Petäjä-Repo; István Szokodi; Risto Kerkelä

The G protein-coupled apelin receptor regulates important processes of the cardiovascular homeostasis, including cardiac development, cardiac contractility, and vascular tone. Most recently, a novel endogenous peptide ligand for the apelin receptor was identified in zebrafish, and it was named apela/elabela/toddler. The peptide was originally considered as an exclusively embryonic regulator, and so far its function in the adult organism remains elusive. We show here that apela is predominantly expressed in the non-cardiomyocyte fraction in the adult rodent heart. We also provide evidence that apela binds to apelin receptors in the heart. Using isolated adult rat hearts, we demonstrate, that just like the fellow receptor agonist apelin, apela increases cardiac contractility and induces coronary vasodilation already in the nanomolar level. The inotropic effect, as revealed by Western blot analysis, is accompanied by a significant increase in extracellular signal-regulated kinase (ERK) 1/2 phosphorylation. Pharmacological inhibition of ERK1/2 activation markedly attenuates the apela-induced inotropy. Analysis of samples from infarcted mouse hearts showed that expression of both apela and apelin receptor is induced in failing mouse hearts and correlate with left ventricular ejection fraction. Hence, we conclude that apela is present in the adult heart, is upregulated in post-infarction cardiac remodeling, and increases cardiac contractility in an ERK1/2-dependent manner.


Acta Physiologica | 2012

Physiological regulation of cardiac contractility by endogenous reactive oxygen species

Ábel Perjés; Anna Maria Kubin; Attila Kónyi; S. Szabados; A. Cziráki; Reka Skoumal; Heikki Ruskoaho; István Szokodi

Increased production of reactive oxygen species (ROS) has been linked to the pathogenesis of congestive heart failure. However, emerging evidence suggests the involvement of ROS in the regulation of various physiological cellular processes in the myocardium. In this review, we summarize the latest findings regarding the role of ROS in the acute regulation of cardiac contractility. We discuss ROS‐dependent modulation of the inotropic responses to G protein‐coupled receptor agonists (e.g. β‐adrenergic receptor agonists and endothelin‐1), the potential cellular sources of ROS (e.g. NAD(P)H oxidases and mitochondria) and the proposed end‐targets and signalling pathways by which ROS affect contractility. Accumulating new data supports the fundamental role of endogenously generated ROS to regulate cardiac function under physiological conditions.


Journal of Molecular and Cellular Cardiology | 2014

p38α regulates SERCA2a function.

Leena Kaikkonen; Johanna Magga; Veli Pekka Ronkainen; Elina Koivisto; Ábel Perjés; J. Kurt Chuprun; Leif Erik Vinge; Teemu Kilpiö; Jani Aro; Johanna Ulvila; Tarja Alakoski; James A. Bibb; István Szokodi; Walter J. Koch; Heikki Ruskoaho; Risto Kerkelä

cAMP-dependent protein kinase (PKA) regulates the L-type calcium channel, the ryanodine receptor, and phospholamban (PLB) thereby increasing inotropy. Cardiac contractility is also regulated by p38 MAPK, which is a negative regulator of cardiac contractile function. The aim of this study was to identify the mechanism mediating the positive inotropic effect of p38 inhibition. Isolated adult and neonatal cardiomyocytes and perfused rat hearts were utilized to investigate the molecular mechanisms regulated by p38. PLB phosphorylation was enhanced in cardiomyocytes by chemical p38 inhibition, by overexpression of dominant negative p38α and by p38α RNAi, but not with dominant negative p38β. Treatment of cardiomyocytes with dominant negative p38α significantly decreased Ca(2+)-transient decay time indicating enhanced sarco/endoplasmic reticulum Ca(2+)-ATPase function and increased cardiomyocyte contractility. Analysis of signaling mechanisms involved showed that inhibition of p38 decreased the activity of protein phosphatase 2A, which renders protein phosphatase inhibitor-1 phosphorylated and thereby inhibits PP1. In conclusion, inhibition of p38α enhances PLB phosphorylation and diastolic Ca(2+) uptake. Our findings provide evidence for novel mechanism regulating cardiac contractility upon p38 inhibition.


Journal of Biological Chemistry | 2012

Neuronostatin, a novel peptide encoded by somatostatin gene, regulates cardiac contractile function and cardiomyocyte survival.

Laura Vainio; Ábel Perjés; Niilo R. I. Ryti; Johanna Magga; Tarja Alakoski; Raisa Serpi; Leena Kaikkonen; Jarkko Piuhola; István Szokodi; Heikki Ruskoaho; Risto Kerkelä

Background: Neuronostatin peptide is a somatostatin gene derivative, whose functions are largely unknown. Results: Neuronostatin activates p38 MAPK and JNK attenuating endothelin-1-induced cardiac contractility and compromising cardiomyocyte viability. Conclusion: Neuronostatin has multiple biological effects in cardiomyocytes. Significance: Receptors for neuronostatin need to be identified to further characterize the functions of the peptide. Neuronostatin, a recently discovered peptide encoded by somatostatin gene, is involved in regulation of neuronal function, blood pressure, food intake, and drinking behavior. However, the biological effects of neuronostatin on cardiac myocytes are not known, and the intracellular signaling mechanisms induced by neuronostatin remain unidentified. We analyzed the effect of neuronostatin in isolated perfused rat hearts and in cultured primary cardiomyocytes. Neuronostatin infusion alone had no effect on left ventricular (LV) contractile function or on isoprenaline- or preload-induced increase in cardiac contractility. However, infusion of neuronostatin significantly decreased the positive inotropic response to endothelin-1 (ET-1). This was associated with an increase in phosphorylation of p38 mitogen-activated protein kinase and c-Jun N-terminal kinase (JNK). Treatment of both neonatal and adult cardiomyocytes with neuronostatin resulted in reduced cardiomyocyte viability. Inhibition of JNK further increased the neuronostatin-induced cell death. We conclude that neuronostatin regulates cardiac contractile function and cardiomyocyte survival. Receptors for neuronostatin need to be identified to further characterize the biological functions of the peptide.


Regulatory Peptides | 2010

Prolactin-releasing peptide regulates cardiac contractility

Attila Kónyi; Reka Skoumal; Anna Maria Kubin; Gábor Füredi; Ábel Perjés; Klára Farkasfalvi; Zsolt Sárszegi; Ferenc Horkay; Iván G. Horváth; Miklós Tóth; Heikki Ruskoaho; István Szokodi

High levels of specific prolactin-releasing peptide (PrRP) binding sites have been found in the myocardium; however, the functional importance of PrRP in the regulation of cardiac function is unknown. In isolated perfused rat hearts, infusion of PrRP (1-100 nM) induced a dose-dependent positive inotropic effect. Inhibition of cAMP catabolism by IBMX, a phosphodiesterase inhibitor, failed to augment the contractile effect of PrRP. The protein phosphatase (PP1/PP2A) inhibitor calyculin A increased the inotropic response to PrRP, whereas the PP2A inhibitor okadaic acid had no effect. Ro32-0432, a protein kinase C alpha (PKC alpha) inhibitor, significantly enhanced the inotropic effect of PrRP as well as the phosphorylation of phospholamban at Ser-16. In conclusion, the present data define a hitherto unrecognized role for PrRP in the regulation of cardiovascular system by showing that PrRP exerts a direct positive inotropic effect. Moreover, our results suggest that the cAMP-independent inotropic response to PrRP is suppressed by concurrent activation of PKC alpha and PP1.


Orvosi Hetilap | 2015

Role of reactive oxygen species in the regulation of cardiac function

Iván G. Horváth; Bálint Kittka; Ábel Perjés; Heikki Ruskoaho; István Szokodi

Increased production of reactive oxygen species has been implicated in the pathogenesis of congestive heart failure. However, emerging evidence suggests a role for reactive oxygen species in regulating various physiological cellular processes in the myocardium. The authors summarize the current understanding of involvement of reactive oxygen species in the regulation of cardiac contractility under physiological conditions.Increased production of reactive oxygen species has been implicated in the pathogenesis of congestive heart failure. However, emerging evidence suggests a role for reactive oxygen species in regulating various physiological cellular processes in the myocardium. The authors summarize the current understanding of involvement of reactive oxygen species in the regulation of cardiac contractility under physiological conditions.


Handbook of Biologically Active Peptides (Second Edition) | 2013

Chapter 187 – Apelin

Ábel Perjés; Klara Farkasfalvi; Heikki Ruskoaho; István Szokodi


Cardiologia Croatica | 2014

Signaling mechanisms mediating the positive inotropic response to apelin in the intact rat heart

Peter Ezer; Ábel Perjés; Réka Skoumal; Mihály Simon; Attila Cziraki; Akos Koller; Attila Kónyi; Iván Horváth; Heikki Ruskoaho; Istvan Szokodi

Collaboration


Dive into the Ábel Perjés's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge