Network


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

Hotspot


Dive into the research topics where Eirik Qvigstad is active.

Publication


Featured researches published by Eirik Qvigstad.


Physiological Genomics | 2012

Cardiac O-GlcNAc signaling is increased in hypertrophy and heart failure.

Ida G. Lunde; Jan Magnus Aronsen; Heidi Kvaløy; Eirik Qvigstad; Ivar Sjaastad; Theis Tønnessen; Geir Christensen; Line M. Grønning-Wang; Cathrine R. Carlson

Reversible protein O-GlcNAc modification has emerged as an essential intracellular signaling system in several tissues, including cardiovascular pathophysiology related to diabetes and acute ischemic stress. We tested the hypothesis that cardiac O-GlcNAc signaling is altered in chronic cardiac hypertrophy and failure of different etiologies. Global protein O-GlcNAcylation and the main enzymes regulating O-GlcNAc, O-GlcNAc transferase (OGT), O-GlcNAcase (OGA), and glutamine-fructose-6-phosphate amidotransferase (GFAT) were measured by immunoblot and/or real-time RT-PCR analyses of left ventricular tissue from aortic stenosis (AS) patients and rat models of hypertension, myocardial infarction (MI), and aortic banding (AB), with and without failure. We show here that global O-GlcNAcylation was increased by 65% in AS patients, by 47% in hypertensive rats, by 81 and 58% post-AB, and 37 and 60% post-MI in hypertrophic and failing hearts, respectively (P < 0.05). Noticeably, protein O-GlcNAcylation patterns varied in hypertrophic vs. failing hearts, and the most extensive O-GlcNAcylation was observed on proteins of 20-100 kDa in size. OGT, OGA, and GFAT2 protein and/or mRNA levels were increased by pressure overload, while neither was regulated by myocardial infarction. Pharmacological inhibition of OGA decreased cardiac contractility in post-MI failing hearts, demonstrating a possible role of O-GlcNAcylation in development of chronic cardiac dysfunction. Our data support the novel concept that O-GlcNAc signaling is altered in various etiologies of cardiac hypertrophy and failure, including human aortic stenosis. This not only provides an exciting basis for discovery of new mechanisms underlying pathological cardiac remodeling but also implies protein O-GlcNAcylation as a possible new therapeutic target in heart failure.


Circulation Research | 2005

Dual Serotonergic Regulation of Ventricular Contractile Force Through 5-HT2A and 5-HT4 Receptors Induced in the Acute Failing Heart

Eirik Qvigstad; Ivar Sjaastad; Trond Brattelid; Caroline Nunn; Fredrik Swift; Jon Arne Kro Birkeland; Kurt A. Krobert; Geir Øystein Andersen; Ole M. Sejersted; Jan-Bjørn Osnes; Finn Olav Levy; Tor Skomedal

Cardiac responsiveness to neurohumoral stimulation is altered in congestive heart failure (CHF). In chronic CHF, the left ventricle has become sensitive to serotonin because of appearance of Gs-coupled 5-HT4 receptors. Whether this also occurs in acute CHF is unknown. Serotonin responsiveness may develop gradually or represent an early response to the insult. Furthermore, serotonin receptor expression could vary with progression of the disease. Postinfarction CHF was induced in male Wistar rats by coronary artery ligation with nonligated sham-operated rats as control. Contractility was measured in left ventricular papillary muscles and mRNA quantified by real-time reverse-transcription PCR. Myosin light chain-2 phosphorylation was determined by charged gel electrophoresis and Western blotting. Ca2+ transients in CHF were measured in field stimulated fluo-4-loaded cardiomyocytes. A novel 5-HT2A receptor-mediated inotropic response was detected in acute failing ventricle, accompanied by increased 5-HT2A mRNA levels. Functionally, this receptor dominated over 5-HT4 receptors that were also induced. The 5-HT2A receptor-mediated inotropic response displayed a triphasic pattern, shaped by temporally different activation of Ca2+-calmodulin-dependent myosin light chain kinase, Rho-associated kinase and inhibitory protein kinase C, and was accompanied by increased myosin light chain-2 phosphorylation. Ca2+ transients were slightly decreased by 5-HT2A stimulation. The acute failing rat ventricle is, thus, dually regulated by serotonin through Gq-coupled 5-HT2A receptors and Gs-coupled 5-HT4 receptors.


Cardiovascular Research | 2010

Natriuretic peptides increase β1-adrenoceptor signalling in failing hearts through phosphodiesterase 3 inhibition

Eirik Qvigstad; Lise Román Moltzau; Jan Magnus Aronsen; Cam H.T. Nguyen; Karina Hougen; Ivar Sjaastad; Finn Olav Levy; Tor Skomedal; Jan-Bjørn Osnes

AIMS Whereas natriuretic peptides increase cGMP levels with beneficial cardiovascular effects through protein kinase G, we found an unexpected cardio-excitatory effect of C-type natriuretic peptide (CNP) through natriuretic peptide receptor B (NPR-B) stimulation in failing cardiac muscle and explored the mechanism. METHODS AND RESULTS Heart failure was induced in male Wistar rats by coronary artery ligation. Contraction studies were performed in left ventricular muscle strips. Cyclic nucleotides were measured by radio- and enzyme immunoassay. Apoptosis was determined in isolated cardiomyocytes by Annexin-V/propidium iodide staining and phosphorylation of phospholamban (PLB) and troponin I was measured by western blotting. Stimulation of NPR-B enhanced beta1-adrenoceptor (beta1-AR)-evoked contractile responses through cGMP-mediated inhibition of phosphodiesterase 3 (PDE3). CNP enhanced beta1-AR-mediated increase of cAMP levels to the same extent as the selective PDE3 inhibitor cilostamide and increased beta1-AR-stimulated protein kinase A activity, as demonstrated by increased PLB and troponin I phosphorylation. CNP promoted cardiomyocyte apoptosis similar to inhibition of PDE3 by cilostamide, indicative of adverse effects of NPR-B signalling in failing hearts. CONCLUSION An NPR-B-cGMP-PDE3 inhibitory pathway enhances beta(1)-AR-mediated responses and may in the long term be detrimental to the failing heart through mechanisms similar to those operating during treatment with PDE3 inhibitors or during chronic beta-adrenergic stimulation.


Basic Research in Cardiology | 2007

Expression of mRNA encoding G protein-coupled receptors involved in congestive heart failure--a quantitative RT-PCR study and the question of normalisation.

Trond Brattelid; Kristine Tveit; Jon Arne Kro Birkeland; Ivar Sjaastad; Eirik Qvigstad; Kurt A. Krobert; Rizwan I. Hussain; Tor Skomedal; Jan-Bjørn Osnes; Finn Olav Levy

AbstractCongestive heart failure (CHF) induces changes in the neurohumoral system and gene expression in viable myocardium. Several of these genes encode G protein-coupled receptors (GPCRs) involved in mechanisms which compensate for impaired myocardial function. We used real-time quantitative RT-PCR (Q-RT-PCR) to investigate the expression of mRNA encoding 15 different GPCRs possibly involved in CHF, and the effect of normalisation to GAPDH mRNA (GAPDH) or 18S rRNA (18S). CHF was induced in rats by coronary artery ligation, with sham-operated controls (Sham). After 6 weeks, mRNA expression in viable left ventricular myocardium was determined using both 18S and GAPDH as the normalisation standard. An apparent 30% reduction in GAPDH mRNA levels vs. 18S in CHF compared to Sham, although not significant in itself, influenced the interpretation of regulation of other genes.Thus, levels of mRNA encoding receptors for angiotensin II (AT1), endothelin (ETA, ETB) and the muscarinic acetylcholine (mACh) receptor M1 increased significantly in CHF only when normalised to GAPDH. Levels of mRNA encoding the mACh receptors M3 and M4 and the serotonin receptors 5-HT2A and 5-HT4 increased, whereas α1D-adrenoceptor mRNA decreased in CHF irrespective of the normalisation standard. No significant change was detected for M2 and M5 mACh receptors or α1A-, α1B-, β1- or β2-adrenoceptors. Q-RT-PCR is a sensitive and powerful method to monitor changes in GPCR mRNA expression in CHF. However, the normalisation standard used is important for the interpretation of mRNA regulation.


British Journal of Pharmacology | 2007

Effects of treatment with a 5-HT4 receptor antagonist in heart failure

Jon Arne Kro Birkeland; Ivar Sjaastad; Trond Brattelid; Eirik Qvigstad; E.R. Moberg; Kurt A. Krobert; R. Bjørnerheim; Tor Skomedal; Ole M. Sejersted; Jan-Bjørn Osnes; Finn Olav Levy

Positive inotropic responses (PIR) to 5‐hydroxytryptamine (5‐HT) are induced in the left ventricle (LV) in rats with congestive heart failure (CHF); this is associated with upregulation of the Gs‐coupled 5‐HT4 receptor. We investigated whether chronic 5‐HT4 receptor blockade improved cardiac function in CHF rats.


Journal of Biological Chemistry | 2008

Cardiac-restricted Expression of the Carboxyl-terminal Fragment of GRK3 Uncovers Distinct Functions of GRK3 in Regulation of Cardiac Contractility and Growth GRK3 CONTROLS CARDIAC α1-ADRENERGIC RECEPTOR RESPONSIVENESS

Leif Erik Vinge; Thomas G. von Lueder; Ellen Aasum; Eirik Qvigstad; Jøergen A. Gravning; Ole-Jakob How; Thor Edvardsen; Reidar Bjørnerheim; M. Shakil Ahmed; Birthe W. Mikkelsen; Erik Øie; Toril Attramadal; Tor Skomedal; Otto A. Smiseth; Walter J. Koch; Terje S. Larsen; Håvard Attramadal

G protein-coupled receptor kinase-2 and -3 (GRK2 and GRK3) in cardiac myocytes catalyze phosphorylation and desensitization of different G protein-coupled receptors through specificity controlled by their carboxyl-terminal pleckstrin homology domain. Although GRK2 has been extensively investigated, the function of cardiac GRK3 remains unknown. Thus, in this study cardiac function of GRK3 was investigated in transgenic (Tg) mice with cardiac-restricted expression of a competitive inhibitor of GRK3, i.e. the carboxyl-terminal plasma membrane targeting domain of GRK3 (GRK3ct). Cardiac myocytes from Tg-GRK3ct mice displayed significantly enhanced agonist-stimulated α1-adrenergic receptor-mediated activation of ERK1/2 versus cardiac myocytes from nontransgenic littermate control (NLC) mice consistent with inhibition of GRK3. Tg-GRK3ct mice did not display alterations of cardiac mass or left ventricular dimensions compared with NLC mice. Tail-cuff plethysmography of 3- and 9-month-old mice revealed elevated systolic blood pressure in Tg-GRK3ct mice versus control mice (3-month-old mice, 136.8 ± 3.6 versus 118.3 ± 4.7 mm Hg, p < 0.001), an observation confirmed by radiotelemetric recording of blood pressure of conscious, unrestrained mice. Simultaneous recording of left ventricular pressure and volume in vivo by miniaturized conductance micromanometry revealed increased systolic performance with significantly higher stroke volume and stroke work in Tg-GRK3ct mice than in NLC mice. This phenotype was corroborated in electrically paced ex vivo perfused working hearts. However, analysis of left ventricular function ex vivo as a function of increasing filling pressure disclosed significantly reduced (dP/dt)min and prolonged time constant of relaxation (τ) in Tg-GRK3ct hearts at elevated supraphysiological filling pressure compared with control hearts. Thus, inhibition of GRK3 apparently reduces tolerance to elevation of preload. In conclusion, inhibition of cardiac GRK3 causes hypertension because of hyperkinetic myocardium and increased cardiac output relying at least partially on cardiac myocyte α1-adrenergic receptor hyper-responsiveness. The reduced tolerance to elevation of preload may cause impaired ability to withstand pathophysiological mechanisms of heart failure.


American Journal of Physiology-heart and Circulatory Physiology | 2011

Mechanism of prolonged electromechanical delay in late activated myocardium during left bundle branch block

Kristoffer Russell; Otto A. Smiseth; Ola Gjesdal; Eirik Qvigstad; Per Andreas Norseng; Ivar Sjaastad; Anders Opdahl; Helge Skulstad; Thor Edvardsen; Espen W. Remme

During left bundle branch block (LBBB), electromechanical delay (EMD), defined as time from regional electrical activation (REA) to onset shortening, is prolonged in the late-activated left ventricular lateral wall compared with the septum. This leads to greater mechanical relative to electrical dyssynchrony. The aim of this study was to determine the mechanism of the prolonged EMD. We investigated this phenomenon in an experimental LBBB dog model (n = 7), in patients (n = 9) with biventricular pacing devices, in an in vitro papillary muscle study (n = 6), and a mathematical simulation model. Pressures, myocardial deformation, and REA were assessed. In the dogs, there was a greater mechanical than electrical delay (82 ± 12 vs. 54 ± 8 ms, P = 0.002) due to prolonged EMD in the lateral wall vs. septum (39 ± 8 vs.11 ± 9 ms, P = 0.002). The prolonged EMD in later activated myocardium could not be explained by increased excitation-contraction coupling time or increased pressure at the time of REA but was strongly related to dP/dt at the time of REA (r = 0.88). Results in humans were consistent with experimental findings. The papillary muscle study and mathematical model showed that EMD was prolonged at higher dP/dt because it took longer for the segment to generate active force at a rate superior to the load rise, which is a requirement for shortening. We conclude that, during LBBB, prolonged EMD in late-activated myocardium is caused by a higher dP/dt at the time of activation, resulting in aggravated mechanical relative to electrical dyssynchrony. These findings suggest that LV contractility may modify mechanical dyssynchrony.


British Journal of Pharmacology | 2014

Inhibition of phosphodiesterase‐3 by levosimendan is sufficient to account for its inotropic effect in failing human heart

Øivind Ørstavik; Ata Sh; Jon Riise; Christen P. Dahl; Geir Øystein Andersen; Finn Olav Levy; Tor Skomedal; Jan-Bjørn Osnes; Eirik Qvigstad

Levosimendan is known as a calcium sensitizer, although it is also known to inhibit PDE3. We aimed to isolate each component and estimate their contribution to the increased cardiac contractility induced by levosimendan.


British Journal of Pharmacology | 2009

5-HT4-elicited positive inotropic response is mediated by cAMP and regulated by PDE3 in failing rat and human cardiac ventricles

Faraz Afzal; Kjetil Wessel Andressen; Halvor K. Mørk; Jan Magnus Aronsen; Ivar Sjaastad; Christen P. Dahl; Tor Skomedal; Finn Olav Levy; Jan-Bjørn Osnes; Eirik Qvigstad

The left ventricle in failing hearts becomes sensitive to 5‐HT parallelled by appearance of functional Gs‐coupled 5‐HT4 receptors. Here, we have explored the regulatory functions of phosphodiesterases in the 5‐HT4 receptor‐mediated functional effects in ventricular muscle from failing rat and human heart.


Neuropharmacology | 2008

Effects of serotonin in failing cardiac ventricle: Signalling mechanisms and potential therapeutic implications

Finn Olav Levy; Eirik Qvigstad; Kurt A. Krobert; Tor Skomedal; Jan-Bjørn Osnes

Previously, cardioexcitation by serotonin (5-hydroxytryptamine, 5-HT) was believed to be confined to atria in mammals including man, and mediated through 5-HT(4) receptors in pig and man, but 5-HT(2A) receptors in rat. Recent studies, reviewed here, demonstrate that functional 5-HT(4) receptors can be revealed in porcine and human ventricular myocardium during phosphodiesterase inhibition, and that 5-HT(4) receptor mRNA is increased in human heart failure. In rats, functional 5-HT(4) and 5-HT(2A) receptors appear in the cardiac ventricle during heart failure and mediate inotropic responses through different mechanisms. 5-HT(2A) receptor signalling resembles that from alpha(1)-adrenoceptors and causes inotropic effects through increased myosin light chain phosphorylation, resulting in Ca(2+) sensitisation. 5-HT(4) receptor signalling resembles that from beta-adrenoceptors and causes inotropic effects through a pathway involving cAMP and PKA-mediated phosphorylation of proteins involved in Ca(2+) handling, resulting in enhanced contractility through increased Ca(2+) availability. Cyclic AMP generated through 5-HT(4) receptor stimulation seems more efficiently coupled to increased contractility than cAMP generated through beta-adrenoceptor stimulation. Increasing contractility through cAMP is considered less energy efficient than Ca(2+) sensitisation and this may be one reason why beta-adrenoceptor antagonism is beneficial in heart failure patients. Treatment of heart failure rats with the 5-HT(4) antagonist SB207266 (piboserod) resulted in potentially beneficial effects, although small. Further studies are needed to clarify if such treatment will be useful for patients with heart failure.

Collaboration


Dive into the Eirik Qvigstad's collaboration.

Top Co-Authors

Avatar

Tor Skomedal

Oslo University Hospital

View shared research outputs
Top Co-Authors

Avatar

Finn Olav Levy

Oslo University Hospital

View shared research outputs
Top Co-Authors

Avatar

Ivar Sjaastad

Oslo University Hospital

View shared research outputs
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