PhD School of Heart Research 4th Annual CHFR Symposium PROGRAM Thursday October 12, 2006 08.00 - 16.50 Holmenkollen Park Hotel Rica Oslo, Norway 2 Welcome to the 4th Annual CHFR Symposium Center for Heart Failure Research (CHFR) constitutes a branch of medical research at the Faculty of Medicine at the University of Oslo, Norway. It is also supported by funding from the Eastern Norway Regional Health Authority (Helse Øst). The center represents a concerted action of groups with complementary expertise that combines state-of-the-art gene technology and capabilities to investigate protein function in experimental systems. The systems range from isolated cells and multicellular preparations to integrative physiology in pathophysiological models of heart failure. The clinical research groups of the Center for Heart Failure Research allow for testing of hypotheses in prospective studies of patients. Our main aim is to promote and integrate high quality research from bench to bedside in the field of heart failure. The CHFR Annual Symposium is a yearly event where ongoing scientific work at the Center for Heart Failure Research is presented. We invite national and international speakers to present their recent results. The aim is to establish new collaborations within the CHFR and with national and international researchers outside the CHFR. 3 PROGRAM 08.00 08.20 Registration Welcome by Geir Christensen (UUH) Scientific session I: Myocardial function in heart failure Chairmen: Ole M. Sejersted (UUH) and Halfdan Ihlen (RUH) 08.30 David Eisner, University of Manchester, United Kingdom Calcium and the heart: in health and disease 09.00 Gerd Hasenfuss, University of Göttingen, Germany Calcium cycling in heart failure 09.30 Øyvind Ellingsen, Norwegian University of Science and Technology, Trondheim, Norway Exercise-induced hypertrophy and reverse remodeling: from genes to patients 10.00 Moderated poster session I: Inflammatory mediators in heart failure Moderators: Torbjørn Omland, Akershus University Hospital and Theis Tønnessen (UUH) Moderated poster session II: Receptor-mediated signaling and novel therapies of heart failure Moderators: Jan-Bjørn Osnes (UoO) and Reidar Bjørnerheim (UUH) Moderated poster sessions III: Cardiac and skeletal muscle dysfunction in heart failure Moderators: Ivar Sjaastad (UUH) and Geir Øystein Andersen (UUH) Moderated poster sessions IV: Cardiac and skeletal muscle dysfunction in heart failure Moderator: Jostein Hallén (Norwegian School of Sport Sciences) 11.30 Lunch Scientific session II: Inflammation in heart failure Chairmen: Pål Aukrust (RUH) and Lars Gullestad (RUH) 12.30 Douglas Mann, Baylor College of Medicine, Houston, USA Immunmodulation in heart failure: past, present and future 13.15 Jan Kristian Damås, Rikshospitalet University Hospital, Oslo, Norway Chemokines as targets for treatment in heart failure 13.45 Break 4 Scientific session III: Receptor-mediated signaling and novel therapies of heart failure Chairmen: Håvard Attramadal (RUH) and Arne Westheim (UUH) 14.05 Walter J. Koch, Jefferson Medical College, Philadelphia, USA GRK2 as a potential therapeutic target and biomarker for heart failure 14.50 Break 15.00 Pieter Doevendans, University Medical Center Utrecht, the Netherlands Human cardiac progenitor cells for myocardial repair 15.30 Kenneth Dickstein, University of Bergen, Norway Ongoing clinical trials in heart failure with drugs and devices 16.00 Break Scientific session IV: Diagnostic strategies in heart failure Chairman: Dan Atar, Aker University Hospital 16.20 Peter Buser, University Hospital Basel, Switzerland The contribution of cardiac magnetic resonance imaging in the evaluation of patients with heart failure: state of the art and future developments 16.50 End of symposium UUH RUH UoO Ullevål University Hospital Rikshospitalet University Hospital University of Oslo 5 6 ABSTRACTS 7 Moderated poster session I: Inflammatory mediators in heart failure P01 TNF induces anemia in mice with ischemic heart failure, while NO represses erythropoiesis in non-ischemic heart failure Per O Iversen, Kristin B Andersson, Alexandra V Finsen, Ivar Sjaastad, Thomas von Lueder, Ole M Sejersted, Håvard Attramadal, Geir Christensen P02 Myocardial expression of the TNF superfamily cytokine LIGHT and its receptors in experimental and clinical heart failure Christen P Dahl, Lars Gullestad, Erik Øie, Jan Kristian Damås, Pål Aukrust, Arne Yndestad P03 Elevated levels of activin A and follistatin in clinical and experimental pulmonary arterial hypertension. Arne Yndestad, Karl-Otto Larsen, Erik Øie, Thor Ueland, Camilla Smith, Bente Halvorsen, Ivar Sjaastad, Ole Henning Skjønsberg, Turid M. Pedersen, OleGunnar Anfinsen, Jan Kristian Damås, Geir Christensen, Pål Aukrust, Arne K. Andreassen P04 Leukaemia inhibitory factor stimulates glucose transport in isolated cardiomyocytes and induces insulin resistance after chronic exposure Geir Florholmen, G. Hege Thoresen, Arild C Rustan, Jørgen Jensen, Geir Christensen, Vigdis Aas P05 Connective tissue growth factor inhibits myocardial growth, stimulates fibrosis, but preserves myocardial function in chronic pressure overload M. Shakil Ahmed, Thomas von Lueder, Jørgen A. Gravning, Thor Edvardsen, Erik Øie, Birthe Mikkelsen, Otto A.Smiseth, Håvard Attramadal P06 Novel cardioprotective role of connective tissue growth factor in ischemia/reperfusion injury and heart failure Jørgen A. Gravning, M. Shakil Ahmed, Vladimir Martinov, Thomas G. von Lueder, Gabor Czibik, Thor Edvardsen, Birthe V. Mikkelsen, Otto A. Smiseth, Guro Valen, Håvard Attramadal P07 Plasma chromogranin A levels in the subacute phase are predictive of exercise performance after myocardial infarction Helge Røsjø, Kenneth Dickstein, Torbjørn Omland 8 P01 TNF induces anemia in mice with ischemic heart failure, while NO represses erythropoiesis in non-ischemic heart failure Per O Iversen1, Kristin B Andersson2, Alexandra V Finsen2, Ivar Sjaastad2,3, Thomas von Lueder4, Ole M Sejersted2, Håvard Attramadal4, Geir Christensen2 Department of Nutrition, IMB, University of Oslo Institute for Experimental Medical Research, Ullevaal University Hospital, Oslo 3 Department of Cardiology, Ullevaal University Hospital, Oslo 4 Institute of Surgical Research, Rikshospitalet-Radiumhospitalet Medical Center, Oslo 1 2 Background: Anemia is an independent predictor of poor outcome in patients with congestive heart failure (CHF). Insufficient erythropoietin production due to impaired renal function cannot fully explain CHF-related anemia. In myocardial infarction (MI), the most common cause of CHF, it is thought that a low-grade inflammatory response induced by the infarction itself might inhibit blood cell formation. Aims: We wanted to examine (i) whether anemia would develop in mouse models of non-ischemic CHF, and (ii) to identify possible inhibitors of bone marrow hematopoiesis in CHF. Methods: We examined hematopoiesis in adult mice (n=6-9 per group) with CHF induced by MI (ischemia), and in two genetic mouse models of CHF induced by altered sarcoplasmic reticulum (SR) function (non-ischemia), namely cardiacrestricted overexpression of the Ca2+-binding protein calsequestrin (CSQ), or cardiac-restricted knockout of the SR Ca2+-ATPase 2 gene (SERCA2 KO). Hematopoiesis was analyzed with colony assays. Results: In control mice Hb was 14.1±0.1 g/dl, while it was reduced to 10.1±0.1; 9.7±0.2 and 9.6±0.1 g/dl in MI, CSQ and SERCA2 KO, respectively (P<0.05 for all groups; mean±SEM). Colony numbers of immature hematopoietic progenitors in the three CHF groups were reduced to 33±4 (MI), 39±2 (CSQ) and 32±4 (SERCA2 KO) (P<0.05 for all groups) compared with 71±5 in controls. In MImice, plasma TNF and TNF mRNA levels among bone marrow T cells increased 2-4 fold, and anti-TNF antibodies fully restored growth of those cells. However, no such changes were observed in CSQ or SERCA2 KO compared with control mice. In contrast, inhibition of hematopoiesis could be nearly completely abolished with eNOS-blockade in CSQ and SERCA2 KO, but not in MI-mice. Conclusions: This study demonstrates different mechanisms of anemia in CHF; whereas TNF can impair hematopoiesis in MI-induced CHF, NO apparently is a negative regulator of blood cell formation in non-ischemic CHF. 9 P02 Myocardial expression of the TNF superfamily cytokine LIGHT and its receptors in experimental and clinical heart failure Christen P Dahl, Lars Gullestad, Erik Øie, Jan Kristian Damås, Pål Aukrust, Arne Yndestad Institutt for indremedisinsk forskning, Rikshospitalet Purpose: LIGHT (TNFSF14) is an inflammatory cytokine in the tumor necrosis factor (TNF) superfamily that is involved in innate and adaptive immune responses as well as in regulation of cell survival and proliferation. We have previously shown that patients with heart failure (HF) have elevated gene expression of LIGHT in peripheral blood mononuclear cells. In the present study, we examined the myocardial expression of LIGHT and its receptors lymphotoxin-receptor (LTR) and herpes virus entry mediator (HVEM) in experimental and clinical HF. Methods and results: Myocardial gene expression of LIGHT, LTR, and HVEM was analysed by real-time RT-PCR in a rat model of post-infarction HF 2, 7, and 28 days after induction of myocardial infarction (MI). LIGHT mRNA levels were markedly elevated in the infarcted area and modestly increased in the nonischemic part of left ventricle throughout the study period. These changes in LIGHT were accompanied by increased expression of its corresponding receptors of which LTR was moderately increased at all time points after MI, while HVEM gene expression reached the highest levels 28 days post-MI, representing a chronic stage of HF. Immunohistochemical analysis of left ventricular tissue from explanted failing human hearts demonstrated fairly strong LIGHT and HVEM immunoreactivity in cardiomyocytes, endothelial cells, and vascular smooth muscle cells, whereas weaker LTR immunoreactivity was observed in cardiomyocytes. Conclusion: Based on the important role of LIGHT in regulation of the immune response, our findings in both clinical and experimental HF, suggest a role for myocardial autocrine/paracrine LIGHT signalling pathways in the pathogenesis of HF. 10 P03 Elevated levels of activin A and follistatin in clinical and experimental pulmonary arterial hypertension. Arne Yndestad1, Karl-Otto Larsen2,3,4, Erik Øie5,6, Thor Ueland1,7, Camilla Smith1, Bente Halvorsen1, Ivar Sjaastad2,4,8, Ole Henning Skjønsberg3, Turid M. Pedersen1, Ole-Gunnar Anfinsen9, Jan Kristian Damås1,10, Geir Christensen2,4, Pål Aukrust1,10, Arne K. Andreassen4,9 Research Institute for Internal Medicine, Rikshospitalet University Hospital Institute for Experimental Medical Research, Ullevål University Hospital 3 Department of Pulmonary Medicine, Ullevål University Hospital 4 Center for Heart Failure Research 5 Institute for Surgical Research, Rikshospitalet University Hospital 6 Medical Department, Diakonhjemmet Hospital 7 Department of Endocrinology, Rikshospitalet University Hospital 8 Department of Cardiology, Ullevål University Hospital 9 Department of Cardiology 10 Section of Clinical Immunology and Infectious Diseases, Rikshospitalet University Hospital, University of Oslo, Norway. 1 2 Objectives. We sought to explore a role for activin A in pulmonary arterial hypertension (PAH). Background. Growing evidence links the transforming growth factor-beta superfamily to the pathogenesis of PAH. Activin A, a member of this cytokine family, is involved in regulation of cell proliferation, tissue remodeling and inflammation. Methods. Activin A and its binding protein follistatin were measured by enzyme immune assays in mixed venous and arterial blood from patients with PAH [idiopathic PAH (n=18), associated PAH (n=18), and chronic thromboembolic PH (n=14)] and control subjects (n=14) and further explored in experimental studies. Results. i) Levels of activin A and follistatin were significantly elevated in patients with PAH. ii) Activin A levels were significantly related to mortality in the patient cohort. iii) PAH patients had significantly higher follistatin levels in the femoral than in the pulmonary artery indicating net release of follistatin from lungs. iv) Lungs from mice with hypoxia-induced PAH exhibited increased gene expression of activin A and follistatin, and also elevated levels of phosphorylated and total Smad2. v) Immunohistochemistry of human lung autopsies from PAH patients and lungs with experimental PAH localized activin A primarily to alveolar macrophages and bronchial epithelial cells; vi) Activin A induced gene expression of endothelin-1 and plasminogen activator inhibitor (PAI)-1 in endothelial cells and vascular smooth muscle cell, respectively; mediators that both could contribute to vascular remodeling. Conclusion. Our findings in both clinical and experimental studies suggest a role for activin A in the development of PAH. 11 P04 Leukaemia inhibitory factor stimulates glucose transport in isolated cardiomyocytes and induces insulin resistance after chronic exposure Geir Florholmen1,2, G. Hege Thoresen3, Arild C Rustan3, Jørgen Jensen4, Geir Christensen1,2, Vigdis Aas3,5 1 2 3 4 Institute for Experimental Medical Research, Ullevaal University Hospital. Center for Heart Failure Research, University of Oslo. Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo. National Institute of Occupational Health, Oslo. (5)Faculty of Health Sciences, Oslo University College. Hypertrophic and failing hearts have increased utilization of glucose, but develop insulin resistance and reduced ATP production. Increased levels of the interleukin-6-related cytokine leukaemia inhibitory factor (LIF) are found in failing hearts, and we have recently shown that LIF reduces ATP production in isolated cardiomyocytes. In the present study we investigated effects of LIF on glucose metabolism, and how LIF-treated cells respond to insulin stimulation. Cardiomyocytes were isolated from adult Wistar rats and treated with 1 nmol/l LIF. Acute LIF treatment increased deoxyglucose uptake compared to control, but no additive effect was observed in cardiomyocytes treated with both LIF and insulin. The phosphatidylinositol 3-kinase inhibitor wortmannin did not affect the LIF-induced glucose uptake. LIF had no effect on AMPK phosphorylation. Cardiomyocytes treated with LIF for 48h did not respond to insulin by increasing deoxyglucose uptake and showed reduced insulin-mediated uptake of oleic acid and formation of complex lipids compared to control cells. Chronic LIF treatment increased gene expression of the suppressor of cytokine signalling (SOCS) 3 and reduced expression of the glucose transporter GLUT4. In line with these observations, chronic LIF treatment reduced insulin-mediated phosphorylation of both Akt/PKB and glycogen synthase kinase (GSK)-3. Acute LIF treatment increased glucose uptake in isolated cardiomyocytes by another pathway than insulin. Chronic LIF treatment induced insulin resistance, possibly mediated by altered expression of SOCS3 and GLUT4, and impaired insulin-mediated phosphorylation of GSK-3 and Akt/PKB. 12 P05 Connective tissue growth factor inhibits myocardial growth, stimulates fibrosis, but preserves myocardial function in chronic pressure overload M. Shakil Ahmed1, Thomas von Lueder1,2, Jørgen A. Gravning1, Thor Edvardsen1,2, Erik Øie1,2, Birthe Mikkelsen1, Otto A.Smiseth1,2, Håvard Attramadal1,2 1 2 Inst. for Surgical Research and Dept. of Cardiology, Rikshospitalet-Radiumhospitalet Medical Center and University of Oslo. Background: Myocardial expression of connective tissue growth factor (CTGF) is dramatically induced in heart failure (HF) of diverse etiologies. However, the physiologic and pathophysiologic roles of myocardial CTGF remain unresolved. Methods and Results: To elucidate the actions of myocardial CTGF and its putative role in HF transgenic mice with cardiac-restricted overexpression of CTGF were generated. Transgenic CTGF (Tg-CTGF) mice had slightly lower cardiac mass than that of non-transgenic littermate controls (NLC) (heart weight/tibia length of 4 months old male Tg-CTGF vs. NLC; 58.9±2.7 vs. 68.1±1.2 mg/cm, p<0.05). Consistently, echocardiography revealed significantly smaller end-diastolic interventricular septum thickness in Tg-CTGF. Simultaneous LV pressure-volume analysis did not disclose significant alterations of contractility and cardiac output. Histochemical analysis of myocardial tissue revealed interstitial fibrosis in Tg-CTGF mice. Increased myocardial collagen was confirmed by quantitation of myocardial hydroxyproline contents by HPLC (1.10±0.03 vs.0.82±0.05 pmol/mg dry weight in Tg-CTGF vs. NLC mice; p<0.05). Consistently, increased myocardial procollagen--I and -III mRNA levels in TgCTGF vs. NLC hearts were found. Morover, increased expression of antihypertrophic TGF-2 and GDF-15 mRNA, and decreased expression of EGF mRNA in Tg-CTGF vs. NLC mice was detected. Tg-CTGF and NLC mice were subjected to pressure overload by abdominal aortic banding (AB) or shamoperation (SH). Four weeks after AB, significant elevations of cardiac mass were observed both in Tg-CTGF-AB and NLC-AB mice. However, cardiac hypertrophy was significantly diminished in Tg-CTGF-AB versus NLC-AB. Simultaneous PVanalysis provided evidence of cardiac dysfunction in NLC-AB mice, i.e. significantly increased LVEDD, LVEDP, and decreased stroke volume and cardiac output compared to NLC-SH mice. Strikingly, Tg-CTGF-AB revealed essentially preserved LV pressure-volume relations. Elevations of myocardial BNP mRNA expression were significantly attenuated in Tg-CTGF-AB compared to NLC-AB mice. Conclusion: Myocardial CTGF causes interstitial fibrosis, but no evidence of restrictive cardiac dysfunction, and preserves left ventricular function due to pressure overload. 13 P06 Novel cardioprotective role of connective tissue growth factor in ischemia/reperfusion injury and heart failure Jørgen A. Gravning1, M. Shakil Ahmed1, Vladimir Martinov2, Thomas G. von Lueder1, 3, Gabor Czibik2, Thor Edvardsen1,3, Birthe V. Mikkelsen1, Otto A. Smiseth1, 3, Guro Valen2, Håvard Attramadal1, 3 1 2 3 Inst. for Surgical Research and Dept. of Physiology, University of Oslo Dept. of Cardiology, Rikshospitalet-Radiumhospitalet Medical Center, Oslo, Norway Background: The myocardial growth factor CCN2/CTGF - connective tissue growth factor has been shown to be robustly induced in experimental models of heart failure (HF) as well as in human HF. Yet, its role in the pathophysiologic mechanisms of HF is unknown. Methods and Results: To elucidate the role of myocardial CTGF in ischemia/reperfusion injury and HF, transgenic mice with cardiac-restricted overexpression of CTGF were employed. Transgenic CTGF (Tg-CTGF) hearts had slightly lower cardiac mass than that of non-transgenic littermate controls (NLC) (heart weight/tibia length of 4 months old male Tg-CTGF vs. NLC mice; 58.9±2.7 vs. 68.1±1.2 mg/cm, p<0.05). However, cardiac function assessed by LV pressure-volume analysis was essentially unaltered. Global analysis of gene expression by DNA microarray revealed activation of distinct gene programs in Tg-CTGF hearts, including induction of endoplasmatic reticulum stress response genes, antiapoptotic factors, and ECM proteins, suggesting involvement of CTGF in tissue protection. Also, several signalling mediators reported to confer cardioprotection, e.g. GDF-15, Toll-like receptor-4, and HIF-1, were elevated in Tg-CTGF vs. NLC hearts. Langendorff-perfused hearts from Tg-CTGF (n=9) and NLC mice (n=9) were subjected to 40 min of global (no-flow) ischemia and subsequently 60 min of reperfusion. End-point analysis demonstrated markedly diminished infarct size in Tg-CTGF vs. NLC hearts (21.0±3.9 vs. 49.1±6.3 %, p<0.01) and enhanced recovery of LV function (LV developed pressure 81.3±18.6 vs. 40.4±21.7 mmHg, p<0.01). Western blot analysis also revealed increased levels of phosphorylated (activated) Akt/PKB and GSK-3 in Tg-CTGF vs. NLC hearts, demonstrating activation of bona fide cardioprotective signalling pathways. Consistently, Tg-CTGF mice subjected to pressure-overload by abdominal aortic banding displayed less hypertrophy and preserved LV pressurevolume relations compared with banded NLC mice. Conclusion: These data provide novel evidence that CTGF confers cardioprotection by preemptive preconditioning due to reprogramming of gene expression and may act as endogenous survival factor in HF. 14 P07 Plasma chromogranin A levels in the subacute phase are predictive of exercise performance after myocardial infarction Helge Røsjø1, Kenneth Dickstein2, Torbjørn Omland1 1 2 Akershus University Hospital, Lørenskog, Norway Stavanger University Hospital, Stavanger, Norway Introduction: Acute myocardial infarction (AMI) is associated with complex neuroendocrine activation and varying degrees of subsequent functional impairment. Chromogranin A (CgA) is widely distributed throughout the neuroendocrine system and may, due to its long in vivo and in vitro half-life, be an attractive candidate for assessment of neuroendocrine activity. Recently, plasma levels of CgA have been related to the severity of symptoms and prognosis in patients with chronic heart failure. Hypothesis: We hypothesized that CgA levels in the subacute phase would be predictive of exercise performance after AMI and perform better or equally well as traditional neurohormonal markers. Methods: Blood for determination of neurohormonal markers were obtained on day 3 after symptom debut in 73 male patients (median age 69, interquartile range 66-75 yrs) with documented AMI. CgA in plasma was determined by radioimmunoassay, norepinephrine with HPLC and electrochemical detection and BNP with an immunoradiometric method (ShionogiRIA). Functional capacity 6 months post AMI was assessed during symptom-limited exercise on an upright bicycle ergometer, using a continuous ramp protocol with 20 Watt increments per min and expressed as exercise duration. Patients were prospectively subdivided according to the presence or absence of significant ST-segment depression. Results: The correlations between neuroendocrine markers and exercise duration are shown in the table. Conclusion: Plasma CgA obtained in the subacute phase after AMI are predictive of subsequent exercise performance and performs at least as well as tradional neuroendocrine markers. The relationship is particularly pronounced in patients without exercise-induced ischemia. Table. Correlations between neuroendocrine markers and exercise duration All patients No ST ST depression depression Chromogranin A r= –0.39 * r= –0.58 * r= –0.33 † BNP r= –0.41 * r= –0.46 * r= –0.35 † Norepinephrine r= –0.20 r= –0.40 † r= 0.008 * P < 0.01, † P < 0.05 15 Moderated poster sessions II: Receptor-mediated signaling and novel therapies of heart failure P08 Atorvastatin normalizes endothelial function in healthy smokers Stefan Agewall P09 Inhibition of G protein-coupled receptor kinase-3 (GRK3) rescues cardiac dysfunction after pressure overload Thomas G. von Lueder, Jørgen A. Gravning, Thor Edvardsen, Ole Jacob How, Ellen Aasum, Leif E. Vinge, M. Shakil Ahmed, Birthe V. Mikkelsen, Terje S. Larsen, Otto A. Smiseth, Håvard Attramadal P10 The 5-HT4 receptor – a foetal gene reactivated in heart failure? Trond Brattelid, Silje V.S. Bekkevold, Eirik Qvigstad, Lise Román Moltzau, Dagny L. Sandnes, Jon Arne K. Birkeland, Ivar Sjaastad, Finn Olav Levy P11 5-HT4 mediated positive inotropic response is regulated by PDE3 in failing rat heart and human ventricle Faraz Afzal, Kjetil W. Andressen, Halvor K. Mørk, Magnus Aronsen, Ivar Sjaastad, Lise R. Moltzau, Tor Skomedal, Finn Olav Levy, Jan-Bjørn Osnes, Eirik Qvigstad P12 Effects of treatment with a 5-HT4 serotonin receptor antagonist in heart failure Jon Arne Kro Birkeland, Ivar Sjaastad, Trond Brattelid, Eirik Qvigstad, Enrica Ratti Moberg, Kurt A. Krobert, Reidar Bjørnerheim, Tor Skomedal, Ole M. Sejersted, Jan-Bjørn Osnes, Finn Olav Levy P13 Neurohormonal signalling during decompensation in SERCA knock out mice. Karina Hougen, Jan Magnus Aronsen, Jon Arne Birkeland, Eirik Qvigstad, Bill Louch, Kristin Andersson, Geir Christensen, Ole Sejersted, Finn Olav Levy, Ivar Sjaastad P14 Muscarinic receptor activation elicits an inotropic effect in failing rat ventricle through myosin light chain phosphorylation Rizwan I. Hussain, Eirik Qvigstad, Jon Arne Birkeland, Ivar Sjaastad, Annette Glende, Hilde Eikemo, Trond Brattelid, Jan-Bjørn Osnes, Tor Skomedal, Finn Olav Levy, Kurt A. Krobert P15 Evidence for a novel kinase phosphorylating MLC-2 in quiescent cardiomyocytes Hilde Eikemo, Finn Olav Levy, Tor Skomedal, Jan-Bjørn Osnes 16 P08 Atorvastatin normalizes endothelial function in healthy smokers Stefan Agewall1 1 Division of Cardiology, Aker University Hospital Endothelial function predicts cardiovascular disease. The aim of this study was to examine whether endothelial function in smokers with normal cholesterol levels could be normalized by treatment with 80 mg atorvastatin in comparison with a control group. Healthy smokers (n=20) and non-smokers (n=20) were randomized to 80 mg atorvastatin or to placebo in an open, randomized crossover study. Endothelial functional (Flow-mediated dilatation; FMD) was measured. At baseline smokers had a lower FMD compared to the non-smoking group, 2.2 % ± 0.5 and 4.5 % ± 0.9, respectively (p<0.05). In the smoker group FMD increased to 4.0 % ± 0.8 during treatment with atorvastatin (p<0.05) and returned to basal level during placebo, 2.3 % ± 0.6. In the non-smoking group FMD was unaffected by both atorvastatin and placebo. The net change of total cholesterol or LDL cholesterol was not associated with the net change of FMD when the group was considered as a whole or the smoking group was considered separately. In conclusion, an improved endothelial function was observed in a group of smokers, when they were on 80 mg atorvastatin, whereas atorvastatin had no effect on endothelial function in the non-smoking group. The improved FMD among smokers was independent of the LDL decrease during atorvastatin treatment. The results show that poor endothelial function in smokers with normal lipid levels can be improved by a statin treatment. 17 P09 Inhibition of G protein-coupled receptor kinase-3 (GRK3) rescues cardiac dysfunction after pressure overload Thomas G. von Lueder1, 2, Jørgen A. Gravning1, Thor Edvardsen1, 2, Ole Jacob How3, Ellen Aasum3, Leif E. Vinge1, M. Shakil Ahmed1, Birthe V. Mikkelsen1, Terje S. Larsen3, Otto A. Smiseth1, 2, Håvard Attramadal1, 2 1 2 3 Inst. for Surgical Research and Dept. of Cardiology, Rikshospitalet-Radiumhospitalet Medical Center and University of Oslo, and Dept. of Physiology, University of Tromsø, Norway Background: Previous studies of transgenic mice with cardiac-restricted inhibition of GRK3 disclosed increased cardiac performance and phenotype consistent with 1-adrenergic receptor hyperresponsiveness. This is in contrast to GRK2 (ARK1), an isoenzyme with selectivity for -adrenergic receptors. The aim of this study was to investigate the role of GRK3 during evolving cardiac dysfunction after pressure overload. Methods and results: GRK3ct transgenic mice (Tg-GRK3ct) with cardiac-restricted expression of mini-gene encoding the carboxyl-terminal membrane targeting domain of GRK3 were employed. Inhibition of endogenous GRK3 was confirmed by enhanced 1-AR mediated activation of ERK1/2 in cardiac myocytes from TgGRK3ct as compared to non-transgenic littermate controls (NLC). Tg-GRK3ct mice characteristically displayed myocardial hypercontractility despite unaltered cardiac mass compared to NLC. Pressure overload was induced by abdominal aortic banding (AB) in weight-matched male Tg-GRK3ct (GRK3ct-AB) and NLC (NLC-AB) mice and compared with sham-operated Tg-GRK3ct (GRK3ct-SH) and NLC (NLC-SH) mice. Echocardiography revealed similar baseline cardiac dimensions among all groups. Eight and twelve weeks after AB, significant increases of cardiac mass were found in AB mice compared to sham, but no differences between GRK3ct-AB and NLC-AB were discerned. Eight weeks after AB, analysis of electrically paced, ex vivo perfused hearts at increasing filling pressures revealed preserved end-diastolic and end-systolic pressure-volume (PV) relations in Tg-GRK3ct-AB (n=10), with substantially higher cardiac output and dP/dtmax compared to NLC-AB (n=10). Twelve weeks after AB, simultaneous in vivo PV-analysis revealed elevated end-diastolic pressure (10.6±3.1 vs. 2.7±2.2 mmHg, p<0.05) and lower cardiac output (7379±738 vs. 9847±785 μL/min, p<0.05) in NLC-AB (n=11) compared to GRK3ct-AB (n=16). Consistently, myocardial mRNA levels of B-type natriuretic peptide were substantially elevated in NLC-AB vs. GRK3ct-AB (p<0.05). Conclusion: Inhibition of cardiac GRK3 in mice rescues cardiac dysfunction and heart failure after pressure overload. 18 P10 The 5-HT4 receptor – a foetal gene reactivated in heart failure? Trond Brattelid1,2, Silje V.S. Bekkevold1,2, Eirik Qvigstad1,2, Lise Román Moltzau1,2, Dagny L. Sandnes1, Jon Arne K. Birkeland2,3, Ivar Sjaastad2,3,4, Finn Olav Levy1,2 1 2 3 4 Department of Pharmacology, University of Oslo, Oslo, Norway Center for Heart Failure Research, University of Oslo, Oslo, Norway Institute for Experimental Medical Research, University of Oslo, Oslo, Norway Department of Cardiology, Ullevaal University Hospital, Oslo, Norway A positive inotropic responsiveness to serotonin, mediated by 5-HT4 and 5-HT2A receptors, appears in the ventricle of rats with post-infarction congestive heart failure (CHF). A hallmark of CHF is a transition towards a foetal genotype which correlates with loss of cardiac functions. To explore the role of 5-HT4, 5-HT2A and 5-HT2B receptors in the foetal heart, ventricle from Wistar rats were collected 3 and 1 days before expected birth (days -3 and -1), as well as 1, 3, 5 and 113 days after birth, and examined for expression of 5-HT4, 5-HT2A and 5-HT2B serotonin receptor mRNA (real-time quantitative RT-PCR) as well as contractile function mediated by these receptors ex vivo. Both 5-HT4 mRNA expression and function was highest at day -3 and decreased gradually from day -3 to day 5, with a further decrease to adult levels (day 113). The levels increased in CHF, but the levels seen in the foetal ventricle were higher than the levels in CHF. The 5-HT2A and 5-HT2B receptor mRNA levels increased to a maximum immediately after birth, a situation associated with an acute increase in wall stress, but only the 5-HT2A receptor mediated a ventricular inotropic response. These results indicate that the 5-HT4 receptor is a representative of the foetal cardiac gene program and might play a significant role in early cardiac development and CHF. The 5-HT2A and 5-HT2B receptors might therefore play important roles in postnatal heart development associated with an acute increase in wall stress and a subsequent hypertrophy. 19 P11 5-HT4 mediated positive inotropic response is regulated by PDE3 in failing rat heart and human ventricle Faraz Afzal1, Kjetil W. Andressen1, Halvor K. Mørk2, Magnus Aronsen2, Ivar Sjaastad2,3, Lise R. Moltzau1, Tor Skomedal1, Finn Olav Levy1, Jan-Bjørn Osnes1, Eirik Qvigstad1. 1 2 3 4 Department of Pharmacology, University of Oslo Institute for Experimental Medical Research, Ullevål University Hospital Department of Cardiology, Ullevål University Hospital Center for Heart Failure Research, University of Oslo Background: In chronic heart failure, the left ventricle becomes sensitive to serotonin (5-HT) due to the appearance of functional Gs-coupled 5-HT4 receptors. Phosphodiesterase (PDE) 3 and 4 account for >90% of the total cardiac cAMP PDE activity and PDE 4 primarily couples to the cAMP pool activated by adrenoceptors. Objective: Explore the involvement of phosphodiesterases in the regulation of ventricular 5-HT4 mediated positive inotropic effect (PIE) induced in failing rat and human heart. Results: In rat papillary muscles 5-HT4 stimulation exerted a PIE accompanied by an increase in total cAMP and phospholamban phosphorylation. The PIE was increased by a non-selective PDE inhibitor (IBMX, 10µM) and suppressed by the PKA inhibitor H89 (20µM) demonstrating the involvement of the cAMP-PKA pathway. The PDE4 inhibitor rolipram (10µM) did not significantly increase the 5HT4 response. The PDE3 inhibitor cilostamide (1µM) increased the PIE of serotonin without changing the sensitivity. Combined PDE3/4 inhibition further enhanced the PIE and increased the sensitivity to 5-HT. In failing human ventricle PDE3 inhibition significantly increased the PIE whereas PDE4 inhibition had no significant effect. Conclusions: The inotropic response to 5-HT4 is mediated through a cAMP-PKA dependent pathway. PDE3 is the main regulator of this response and the involvement of PDE4 is demasked by inhibition of PDE3. 20 P12 Effects of treatment with a 5-HT4 serotonin receptor antagonist in heart failure Jon Arne Kro Birkeland1, 2, Ivar Sjaastad1, 2, 4, Trond Brattelid1, 3, Eirik Qvigstad1,3, Enrica Ratti Moberg1, 3, Kurt A. Krobert1, 3, Reidar Bjørnerheim1, 4, Tor Skomedal1, 3 , Ole M. Sejersted1, 2, Jan-Bjørn Osnes1, 3, Finn Olav Levy1, 3 1 2 3 4 Center for Heart Failure Research, University of Oslo Institute for Experimental Medical Research, Ullevaal University Hospital Department of Pharmacology, University of Oslo Department of Cardiology, Ullevaal University Hospital We have previously found that a positive inotropic response (PIR) to serotonin (5HT) appears in the left ventricle (LV) of rats with congestive heart failure (CHF), associated with upregulation of the Gs-coupled 5-HT4 receptor. We hypothesized that in CHF, 5-HT4 receptor stimulation could be of disadvantage, as stimulation of beta-adrenoceptors. Therefore, to investigate whether chronic 5-HT4 receptor blockade improves cardiac function in CHF, rats were randomized to receive the 5-HT4 antagonist SB207266 (0.5 mg/kg/24h; MIint; n=30) or placebo (MIpl; n=23) through mini-osmotic pumps for 6 weeks subsequent to induction of postinfarction CHF. LV diastolic function improved, as assessed by a 4.6% lower LV diastolic diameter and 24.2% lower mitral flow deceleration in MIint compared to MIpl (both p<0.05). SB207266 reduced LV systolic diameter by 6.1%, heart weight by 10.2% and lung weight by 13.1% (all p<0.05). The changes in posterior wall thickening and shortening velocity, cardiac output, LV systolic pressure and (dP/dt)max, parameters of LV systolic function, did not reach statistical significance. In isolated papillary muscles, the PIR to isoproterenol (10 µM) increased by 36% and the response to 5-HT (10 µM) decreased by 57% in MIint compared to MIpl (both p<0.05). Real-time quantitative RT-PCR revealed a non-significant reduction in ANP, 5-HT4(b), 5-HT2A and MHCβ mRNA levels and MHCβ/MHC-ratio, and a non-significant increase in MCH mRNA level in MIint compared to MIpl. We conclude that treatment with the 5-HT4 antagonist SB207266 to some extent improved in vivo cardiac function and ex vivo myocardial function, suggesting a possible beneficial effect of treatment with a 5HT4 receptor antagonist in CHF. Since functional 5-HT4 receptors also exist in failing human ventricle, this may be relevant for human heart failure. 21 P13 Neurohormonal signalling during decompensation in SERCA knockout mice Karina Hougen1,3, Jan Magnus Aronsen1,3, Jon Arne Birkeland1,3, Eirik Qvigstad2,3, Bill Louch1,3, Kristin B. Andersson1,3, Geir Christensen1,3, Ole Sejersted1,3, Finn Olav Levy2,3, Ivar Sjaastad1,3,4 1 2 3 4 Institute for Experimental Medical Research, University of Oslo Department of Pharmacology, University of Oslo Center for Heart Failure Research, University of Oslo Department of Cardiology, Ullevaal University Hospital Reduced sarcoplasmic reticulum (SR) function is a hallmark of heart failure. Four weeks after deletion of the SR Ca2+ ATPase (SERCA) gene, mice only exhibit a minor reduction of cardiac function in face of negligible myocardial levels of the SERCA protein. However, these mice develop overt heart failure during the next 3 weeks, although no further reduction in SERCA protein levels are detected. We wanted to examine adrenergic and serotonergic signalling in cardiac myocytes during development of heart failure in these mice. Cardiomyocytes isolated from both SERCA2 KO and control mice 4 and 7 weeks after administration of tamoxifen, were field stimulated at 1Hz. Serotonin receptor (5-HT4 and 5-HT2A) and adrenoceptor mediated effects on fractional cell shortening (FS) was measured in the presence of appropriate receptor blockers present. In a different set of experiments, contractions and Ca2+ transients (fluo4, expressed as F/F0) were measured simultaneously. Fractional shortening in SERCA2 KO cardiomyocytes was only 9.0% of controls at 4 weeks, and 15.7% at 7 weeks (p=NS 4 vs. 7 weeks). In SERCA2 KO cardiomyocytes serotonin increased FS with 56.2 and 88.2% at 4 and 7 weeks respectively. In controls serotonin increased FS with 8.1% and 6.9% respectively. Selective stimulation of 5-HT2A receptors increased FS with 46.9% and F/F0 with 14.9% in SERCA2 KO cardiomyocytes at 7 weeks. In controls FS increased 23.6% and F/F0 increased 3.7%. Isoprenaline increased FS by 284% in SERCA2 KO cardiomyocytes 7 weeks after administration of tamoxifen, compared to 90% in control mice. In conclusion, both serotonergic and adrenergic signaling pathways are functional in SERCA2 KO cardiomyocytes, and are able to enhance cardiomyocyte contractility in vitro.. These responses do not seem to be altered between 4 and 7 weeks after SERCA2 deletion. Thus, other mechanisms might be more important for the observed reduction in vivo function from 4 to 7 weeks. 22 P14 Muscarinic receptor activation elicits an inotropic effect in failing rat ventricle through myosin light chain phosphorylation Rizwan I. Hussain1,2, Eirik Qvigstad1,2, Jon Arne Birkeland1,3, Ivar Sjaastad1,3,4, Annette Glende1,2, Hilde Eikemo1,2, Trond Brattelid1,2, Jan-Bjørn Osnes1,2, Tor Skomedal1,2, Finn Olav Levy1,2, Kurt A. Krobert1,2 1 2 3 4 Center for Heart Failure Research, Faculty of Medicine, University of Oslo Department of Pharmacology, University of Oslo Institute for Experimental Medical Research, Ullevaal University Hospital, University of Oslo Department of Cardiology, Heart and Lung Center, Ullevaal University Hospital Muscarinic receptor mediated inhibition of cAMP dependent inotropic effects is enhanced in papillary muscles from rats with congestive heart failure (CHF). Muscarinic stimulation by the agonist carbachol (CCh) increases myofilament Ca2+ sensitivity without an inotropic effect in normal myocardium. Therefore, we tested the hypothesis that muscarinic activation by CCh could elicit direct inotropic effects in ventricular myocardium from rats with CHF through enhancement of Ca2+ sensitization. Contractile force was measured in papillary muscles from male Wistar rats having undergone ligation of the left coronary artery (and exhibiting CHF) or sham surgery. CCh evoked a positive inotropic effect (20% above basal) approximating 36% of that elicited by isoproterenol (56% above basal), only in rats with CHF. In contrast, CCh elicits only a transient negative inotropic effect in sham rats. The CCh mediated inotropic effect did not correlate with infarction size but did correlate with increased left ventricular end diastolic pressure, heart weight and lung weight, primary indicators of CHF severity. The dose response curve for CCh was shifted to higher concentrations only in the presence of the relatively specific M2 antagonist (AF-DX 116) (pD2values with AF-DX 5.68 ± 0.12 vs. 6.28 ± 0.10 without); whereas, the antagonists nitrocaramiphen (M1) and 4-DAMP (M3/5) were without effect. The CCh mediated inotropic effect was insensitive to pertussis toxin but was attenuated by the myosin light chain kinase inhibitor ML-9 (8% above basal) and by the Rho-kinase inhibitor Y-27632 (9% above basal). CCh increased phosphorylation of myosin light chain that was also attenuated by ML-9 and Y27632. In conclusion, these data indicate that muscarinic receptor activation, possibly M2, mediates an inotropic effect in failing ventricular myocardium, not observed in normal rat heart. The muscarinic inotropic effect occurs through a signaling pathway that enhances myosin light chain phosphorylation and thus, likely Ca2+ sensitization. 23 P15 Evidence for a novel kinase phosphorylating MLC-2 in quiescent cardiomyocytes Hilde Eikemo1,2, Finn Olav Levy1,2, Tor Skomedal1,2, Jan-Bjørn Osnes1,2 1 2 Department of Pharmacology and Center for heart failure research, University of Oslo, Norway Myosin light chain 2 (MLC-2) is a 20 kDa protein located in the hinge region of the heavy myosin molecule. MLC-2 in smooth muscle must be phosphorylated in order to induce contraction. In cardiac muscle, MLC-2 phosphorylation facilitates contraction and is assumed to mediate inotropic responses to various neurohumoral stimuli. The aim of this study was to elucidate some properties of kinases phosphorylating MLC-2 in quiescent cardiomyocytes. Cardiomyocytes were isolated from adult rat hearts by collagenase perfusion and attached to laminine. Phosphorylated and non-phosphorylated MLC-2 were separated using charged gel electrophoresis, and were identified by anti-ventricular MLC-2 monoclonal antibody. The phosphorylation status of MLC-2 is reported as phosphorylated MLC-2 as percent of total MLC-2. Data are presented as mean±SEM. Calyculin A (myosin light chain phosphatase inhibitor) unmasked a kinase activity by increasing MLC-2-phosphorylation dose-dependently from 15.9%±1.8% (0 M) to 45.9%±4.0% (10-7 M), p<0.001, n=6, with a log EC50 of 7.96±0.05, and time-dependently from 18.5%±1.3% at 0 min to 50.1%±3.3% at 80 min, p<0.001, n=12, with half maximal phosphorylation at 15 min and reaching a plateau at about 40 min after addition of calyculin A (10-7 M). Preincubating (45 min) cardiomyocytes with ML-9 (6x10-5 M; myosin light chain kinase (MLCK) and Rho kinase inhibitor) reduced basal MLC-2 phosphorylation, but the effect was reversed by subsequent treatment (time-response curve) with calyculin A (10-7 M). Preincubating with ML-7 (10-5 M; MLCK inhibitor) had no effect, but staurosporine (10-5 M; a broad spectrum protein kinase inhibitor) removed all kinase activity and rendered only a slight effect of calyculin A after 80 min treatment. Conclusion: In quiescent cardiomyocytes, MLC-2 is phosphorylated by a staurosporine sensitive but ML-7 and ML-9 insensitive kinase(s), different from MLCK, probably being Ca2+-independent. 24 25 Moderated poster session III: Cardiac and skeletal muscle dysfunction in heart failure P16 Heart function in adult cardiac Serca2null mice. Kristin Brevik Andersson, Jon Arne Kro Birkeland, Alexandra Vanessa Finsen, William E. Louch, Ivar Sjaastad, Yibin Wang, Ju Chen, Jeffery D. Molkentin, Kenneth R. Chien, Ole M. Sejersted, Geir Christensen P17 Increased transsarcolemmal Ca2+ flux in mice with cardiomyocyte-specific deletion of the Serca2 gene William E. Louch, Jon Arne Kro Birkeland, Alexandra Vanessa Finsen, Kristin Brevik Andersson, Ivar Sjaastad, Yibin Wang, Ju Chen, Jeffery D. Molkentin, Kenneth R. Chien, Ole M. Sejersted, Geir Christensen P18 Role of Serca2 in the embryonic heart Cecilie Sjåland, Geir Christensen, Kristin Brevik Andersson P19 Interplay between the systolic Ca transient, SR Ca content and Ito: a recipe for arrhythmias K.M. Dibb, H.K. Graham, L.C. Diffley, J.D. Clarke, A.W. Trafford P20 Reduced t-tubule density and less synchronous Ca2+ release in cardiomyocytes from rats with congestive heart failure Fredrik Swift, William E. Louch, Jon Arne Kro Birkeland, Marion J. Thomas, Mark B. Cannell, Ivar Sjaastad, Ole M. Sejersted P21 Myocardial infarction in adult mice induces hypertrophy and congestive heart failure without reduced contraction of noninfarcted cardiomyocytes. Halvor K. Mørk, Ivar Sjaastad, Ole M. Sejersted, William E. Louch P22 Differential regulation of genes encoding contractile proteins during development of heart failure after myocardial infarction in mice Cathrine Husberg, Alexandra Finsen, Ståle Nygård, Geir Christensen P23 Calcium handling in cardiomyoctes isolated from rats with post infarction heart failure – effect of hypoxia/reoxygenation and ouabain exposure. Mohammad Nouri Sharikabad, Janne Pedersen, Ivar Sjaastad, Ole M. Sejersted, Odd Brørs P24 Increased amounts of PP1 and PP2A mediating reduced serine 16 phosphorylation of phospholamban is a possible mechanism for cardiac dysfunction in alveolar hypoxia Karl-Otto Larsen, Birgitte Lygren, Ivar Sjaastad, Kurt A. Krobert, Geir Florholmen, Kristin Arnkværn, Ole Henning Skjønsberg, Kjetil Taskén, Geir Christensen 26 P16 Heart function in adult cardiac Serca2null mice. Kristin Brevik Andersson1,2, Jon Arne Kro Birkeland1,2, Alexandra Vanessa Finsen1,2, William E. Louch1,2, Ivar Sjaastad1,2,3, Yibin Wang4, Ju Chen5, Jeffery D. Molkentin6, Kenneth R. Chien7, Ole M. Sejersted1,2, Geir Christensen1,2 1 2 3 4 5 6 7 Institute for Experimental Medical Research, Ullevaal University Hospital Center for Heart Failure Research, University of Oslo Department of Cardiology, Heart and Lung Center, Ullevaal University Hospital Departments of Anesthesiology, Physiology and Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA. Institute of Molecular Medicine, University of California San Diego, La Jolla, California, USA. Department of Pediatrics, University of Cincinnati, Children's Hospital Medical Center, Division of Molecular Cardiovascular Biology, Cincinnati, Ohio, USA. Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, Massachusetts, USA. The generation of Ca2+ transients in cardiomyocytes is essential for heart function. The SERCA2 pump maintains the sarcoplasmic reticulum (SR) Ca2+ store by sequestration of Ca2+ from cytosol into the SR. Loss of SERCA2 in adult hearts is therefore expected to cause immediate severe myocardial contractile dysfunction and death. We have generated a new mouse model to test this hypothesis. Homozygous Serca2flox mice, were mated with Tg(MHC-MerCreMer) transgenic mice. Cardiomyocyte-specific Serca2 deletion may be induced by injection of tamoxifen in adult mice. Tamoxifen-injected Serca2flox/floxTg(MHC-MerCreMer) and control Serca2flox/flox mice are termed SERCA2KO and SERCA2FF mice, respectively. At 4 weeks after tamoxifen induction, the Serca2 gene was efficiently deleted in SERCA2KO myocardium (<3% Serca2 mRNA and <5% SERCA2 protein). Functional thapsigargin-sensitive Ca2+ ATPase was reduced to 15 % of SERCA2FF controls, as measured by 32P-incorporation in tissue homogenates. In isolated cardiomyocytes, we did not detect SERCA2, SERCA1 or SERCA3 proteins. Strikingly, SERCA2KO mice did not present clinical signs of circulatory failure at this timepoint. Contractile parameters were modestly affected, and cardiac output was 80 % of SERCA2 FF control mice. The left atrium was slightly dilated. We found no evidence of hypertrophy, disarray, necrosis or fibrosis in histological sections, nor induction of hypertrophy markers ANP or BNP. However, the lung weight was slightly increased, suggesting some myocardial dysfunction. Protein expression of the L-Ca2+ channel alpha2 regulatory subunit, sodium/calcium exchanger NCX1 and plasma membrane Ca2+-ATPase (PMCA) were all increased in SERCA2KO mice, suggesting alterations in Ca2+ fluxes over the plasma membrane. Cardiomyocyte function in 4 week SERCA2KO mice is presented in the accompanying poster. In conclusion, the SERCA2KO mouse represents a new and unique phenotype of Ca2+-handling in vivo. The model should be useful for mechanistic studies of Ca2+ transport, compensatory mechanisms and progressive heart failure in vivo. 27 P17 Increased transsarcolemmal Ca2+ flux in mice with cardiomyocyte-specific deletion of the Serca2 gene William E. Louch1,2, Jon Arne Kro Birkeland1,2, Alexandra Vanessa Finsen1,2, Kristin Brevik Andersson1,2, Ivar Sjaastad1,2,3, Yibin Wang4, Ju Chen5, Jeffery D. Molkentin6, Kenneth R. Chien7, Ole M. Sejersted1,2, Geir Christensen1,2 1 2 3 4 5 6 7 Institute for Experimental Medical Research, Ullevaal University Hospital, Oslo, Norway. Center for Heart Failure Research, University of Oslo, Oslo, Norway. Department of Cardiology, Heart and Lung Center, Ullevaal University Hospital, Oslo, Norway. Departments of Anesthesiology, Physiology and Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA. Institute of Molecular Medicine, University of California San Diego, La Jolla, California, USA. Department of Pediatrics, University of Cincinnati, Children's Hospital Medical Center, Division of Molecular Cardiovascular Biology, Cincinnati, Ohio, USA. Cardiovascular Research Center, Massachusetts General Hospital, Charlestown, Massachusetts, USA. Heart failure is associated with reduced activity of the sarcoplasmic reticulum (SR) Ca2+ ATPase (SERCA) in cardiomyocytes. To investigate the cardiac phenotype following SERCA downregulation, we recently created a genetically modified mouse which allows conditional cardiomyocyte-specific excision of the Serca2 gene. Four weeks after gene deletion SERCA2 knockout mice (SERCA KO) exhibit very low levels of SERCA2 protein (< 5% control) although the animals do not exhibit signs of heart failure. Here we examined whether endogenous mechanisms compensate for reduction in SERCA expression in isolated left ventricular cardiomyocytes. SERCA KO myocytes exhibited markedly reduced contraction magnitudes relative to flox-flox controls (SERCA FF) during 1 Hz field stimulation (SERCA KO = 27% SERCA FF values, P<0.05). Relaxation was also much slower in SERCA KO cells (maximal relaxation rate = 7% of SERCA FF). Parallel alterations were observed in Ca2+ homeostasis; Ca2+ transients (fluo-4 AM) were markedly smaller in SERCA KO than SERCA FF and the rate of Ca2+ decline was reduced. The releasable SR Ca2+ content was decreased in SERCA KO to 27% of SERCA FF values. Comparison of transients in the absence and presence of caffeine indicated that while SR Ca2+ release contributed to 91% of the Ca2+ transient in SERCA FF, only 14% of the Ca2+ transient in SERCA KO was SR-derived. The peak L-type Ca2+ current was 53% larger in SERCA KO than SERCA FF, and the outward Na+-Ca2+ exchange current assessed during voltage ramps was increased by between 60% (100 mV) and 103% (60 mV). Thus, increased cycling of Ca2+ across the sarcolemma in SERCA KO cells compensates for diminished contribution of the SR to the Ca2+ transient. 28 P18 Role of Serca2 in the embryonic heart Cecilie Sjåland1,2, Geir Christensen1,2, Kristin Brevik Andersson1,2 1 2 Institute for Experimental Medical Research, Ullevaal University Hospital, Oslo, Norway. Center for Heart Failure Research, University of Oslo, Oslo, Norway. The SR Ca2+-ATPase, SERCA2 plays a major role in transporting Ca2+ from the cytosol into the SR during the relaxation state of the excitation-contraction cycle in the heart. Several genetically modified mice have been used to study Serca2 functions in vivo, however homozygous Serca2null die in utero, thus precluding further analysis of Serca2 function. To study the Serca2 function specifically in the heart, we have created the Serca2flox mouse, a genetically modified mouse carrying loxP sites in the Serca2 gene locus Serca2flox such that all Serca2 isoforms would be deleted upon excision mediated by Cre recombinase. Serca2flox mice were mated with MHC-Cre mice in order to generate animals with a heart-specific inactivation of the Serca2 gene from embryonic day 8. Initial breeding results showed a skewed distribution of inheritance of the Serca2flox allele and the MHC-Cre transgene. We found that homozygous knockout Serca2flox/flox Tg(MHC-Cre) embryos died before embryonic day 11.5. This phenotype was unexpected since studies have shown that the Ca2+ transients in the embryonic cardiomyocyte are generate mainly by the L-Ca2+ channel and Na+/ Ca2+-exchanger (NCX1). Furthermore, that the SR is not fully developed until late in embryonic life. We are currently investigating the underlying mechanisms for the unexpected early death of cardiac Serca2 knockout embryos. 29 P19 Interplay between the systolic Ca transient, SR Ca content and Ito: a recipe for arrhythmias K.M. Dibb, H.K. Graham, L.C. Diffley, J.D. Clarke, A.W. Trafford Unit of Cardiac Physiology, The University of Manchester, UK Previously we have shown that the reduction in Ca transient amplitude observed in the hypertrophied heart was brought about by a decrease in SR Ca content. In this study we have investigated if Ca homeostatic mechanisms are altered in a similar way during congestive heart failure (CHF) and how arrhythmogenic risk may be increased despite any reduction in SR Ca content. Heart failure was induced in adult male ferrets by chronic pressure overload of the left ventricle. At the onset of CHF (or the equivalent time period for sham operated animals) myocytes were isolated from the sub-epicardial (epi) and subendocardial (endo) regions of the left ventricle. Experiments were performed using the perforated/whole cell patch technique under current or voltage clamp control at 37°C. Ca transient amplitude was significantly reduced during CHF (680±78 vs 362±51nM sham vs CHF; p<0.05, n=16 to 20). CHF resulted in a reduction in SR Ca content to 76% of sham levels (85±8.6 vs 65±6.2 µM CHF vs sham; p<0.05, n=10 to 23) and we suggest this underpins the reduction in Ca transient amplitude. RT interval (analogous to QT interval) derived from ECG telemetry implants, was found to increase on progression to CHF and ventricular arrhythmias were observed. Action potential duration (90% recovery from peak at 0.5Hz) was increased during CHF in the endo region only (299±25 vs 618±78ms sham vs CHF). The transient outward current (Ito) was decreased in the endo region during CHF (3.0±0.6 vs 1.9±0.5pA/pF sham vs CHF p<0.05). This difference did not exist in the epi region and we suggest that regional alterations in Ito may contribute to the regional changes in action potential duration and subsequently the increased RT interval at CHF. Enhanced transmural gradients in action potential duration and Ito may increase the risk of arrhythmias via re-enterant mechanisms. 30 P20 Reduced t-tubule density and less synchronous Ca2+ release in cardiomyocytes from rats with congestive heart failure Fredrik Swift1, William E. Louch1, Jon Arne Kro Birkeland1, Marion J. Thomas1, Mark B. Cannell2, Ivar Sjaastad1, 3, Ole M. Sejersted1 1 2 3 Institute for Experimental Medical Research and Center for Heart Failure Research, University of Oslo, Ullevaal University Hospital, Oslo, Norway Department of Physiology, Faculty of Medicine and Health Sciences, University of Auckland, New Zealand Department of Cardiology, Heart and Lung Center, Ullevaal University Hospital, Oslo, Norway Release of Ca2+ from the sarcoplasmic reticulum (SR) in cardiomyocytes occurs at functional units where Ca2+ channels located in the t-tubules face SR Ca2+ release channels. The close proximity of these proteins ensure efficient excitationcontraction coupling. Experimentally reducing t-tubule density has been shown to increase the dyssynchrony of Ca2+ release. We investigated whether alterations in t-tubule density and Ca2+ release synchrony occur in cardiomyocytes from rats with post infarction congestive heart failure (CHF). Post infarction CHF was induced by ligation of the left coronary artery. Hearts were fixed with a solution containing 4% paraformaldehyde and mounted in Lowicryl HM20 resin. Thin sections were cut, mounted onto nickel grids, and randomized for blinded examination by electron microscopy. Mean density of the t-tubules was lower in micrographs from failing cardiomyocytes compared to sham (212 t-tubules/100 µm2 vs. 362 t-tubules/100 µm2, p<0.01). T-tubule density was also reduced in isolated cardiomyocytes labeled with Alexa 488 conjugated wheat germ agglutine (WGA). Line-scan images of Ca2+ transients (fluo-4 AM, 1Hz) in field stimulated isolated cardiomyocytes showed that SR Ca2+ release was significantly less synchronous in failing than in sham cells. The degree of dyssynchrony correlated with the severity of CHF. Thus, loss of t-tubules during CHF development may promote dyssynchrony of Ca2+ release, and contribute to decreased efficiency of excitation-contraction coupling in this disease. 31 P21 Myocardial infarction in adult mice induces hypertrophy and congestive heart failure without reduced contraction of noninfarcted cardiomyocytes. Halvor K. Mørk1,2, Ivar Sjaastad1,2,3, Ole M. Sejersted1,2, William E. Louch1 1 2 3 Institute for Experimental Medical Research, Ullevaal University Hospital, Oslo, Norway Center for Heart Failure Research, University of Oslo, Oslo, Norway Department of Cardiology, Heart and Lung Center, Ullevaal University Hospital, Oslo, Norway End-stage heart failure in humans is associated with reduced contraction of individual cardiomyocytes, smaller Ca transients, and reduced Ca content of the sarcoplasmic reticulum. We investigated whether these alterations were present in mice with congestive heart failure (CHF). Myocardial infarction (MI) was induced by ligation of the left coronary artery, and CHF mice were examined at 10-weeks post-MI. SHAM-operated mice served as controls. Heart weight / body weight ratios were 163±20% higher in CHF than SHAM indicating hypertrophy. Lung weight / body weight ratios were also increased (CHF 221±30% of SHAM values) indicating congestion. Echocardiographic analysis showed marked reductions in cardiac output (SHAM 39±4 ml/min; CHF 25±3 ml/min, P<0.05) and left-ventricular fractional shortening (SHAM 27±4%; CHF 8±1%, P<0.05). However, thickening of the non-infarcted posterior wall was unaltered (SHAM 27±6%, CHF 36±7%). Cardiomyocytes isolated from non-infarcted septum were significantly longer in CHF than SHAM (178±8 µm vs 139±6 µm, P<0.05). During field stimulation (1 Hz, 37C), myocyte contraction magnitude was similar in CHF and SHAM cells (% shortening = 9.1±1.1 vs 9.7±0.4, P=NS). Surprisingly, Ca transients (fluo-4 AM) were larger in CHF than in SHAM at both 1Hz (CHF F/F0=3.0±0.2, SHAM F/F0=2.3±0.1, P<0.05) and 5 Hz stimulation frequencies, and the rate of Ca decline was unchanged. Caffeine-elicited transients were of similar magnitude in CHF and SHAM (F/F0=4.3±0.5 vs 5.0±0.4, P=NS), suggesting that sarcoplasmic reticulum Ca content was unaltered. Therefore, CHF progression in mice does not affect contractions of the viable myocardium to the same extent as in heart failure patients. 32 P22 Differential regulation of genes encoding contractile proteins during development of heart failure after myocardial infarction in mice Cathrine Husberg1,2, Alexandra Finsen1,2, Ståle Nygård1,2,3, Geir Christensen1,2 1 2 3 Institute for Experimental Medical Research, Ullevål University Hospital, University of Oslo Center for Heart Failure Research, University of Oslo Department of Mathematics, University of Oslo After myocardial infarction (MI), the non-infarcted region of the left ventricle responds by changing the expression profile of contractile proteins. While the switch from -myosin heavy chain (MHC) to -MHC expression has been well described, the regulation of other proteins in the contractile apparatus has been less studied. We have performed a whole genome profiling of non-infarcted left ventricular tissue isolated from mice at four different time points after left coronary artery ligation. The results from the microarray experiments were evaluated by significance analysis of microarrays (SAM) and the ontological mapping software High-Throughput GoMiner. Significant regulation of several genes encoding proteins involved in muscle development and contraction where identified. Interestingly, the genes encoding signaling molecules (e.g. RhoA, Cdc42, Smpx) showed an early response, suggesting a more rapid reaction to the injury than genes encoding contractile proteins. Among the genes coding for contractile proteins, the most pronounced change was the increased expression of -MHC (16 fold on day 7 post MI). Regarding the various myosin light chain (MLC) encoding genes, a diverse regulation pattern was observed with the highest degree of regulation among regulatory isoforms. Other genes identified as up-regulated where tropomyosin 4 and skeletal muscle -actin, while troponin I was constantly down-regulated. The latter is known to affecting muscle contraction and Ca2+-responsiveness. The results show that whole genome profiling of the non-infarcted left ventricle enables a better understanding of the temporal alterations in the expression of the entire contractile machinery during development of HF. Especially interesting is the extensive alterations in expression of myosin light chains, which may significantly alter the Ca2+responsiveness of the contractile apparatus. 33 P23 Calcium handling in cardiomyoctes isolated from rats with post infarction heart failure – effect of hypoxia/reoxygenation and ouabain exposure. Mohammad Nouri Sharikabad1, Janne Pedersen1, Ivar Sjaastad2, Ole M. Sejersted2, Odd Brørs1 1 2 Clinical Chemistry Department, Ullevål University Hospital, Oslo. Institute for Experimental Medical Research, Ullevål University Hospital, Oslo. Introduction: Knowledge about calcium handling of failing myocardium during hypoxia (low oxygen tension) and reoxygenation is important for understanding the pathology of heart failure and for finding strategies to improve function and survival of the failing myocardium. Aim: The purpose of the present study was to investigate calcium accumulation during reoxygenation of hypoxic cardiomyocytes isolated from rats with postmyocardial-infarction (MI) heart failure compared to control animals. Since influxmode-Na+/Ca2+ exchange is a known candidate for reoxygenation-mediated calcium uptake, the calcium accumulation was also studied in the presence of 1mM ouabain (an inhibitor of Na+/K+ ATPase causing Na+-loading) in normoxic conditions. Methods: Cardiomyocytes were isolated by trypsin/collagenase perfusion of adult Wistar rat hearts from sham-operated animals and animals with post MI heart failure. Hypoxia was induced by changing the atmosphere to 95% nitrogen - 5% CO2 for 4 hours. Cells were then reoxygenated (2 hours) by changing the atmosphere to 95% air - 5% CO(2) and adding glucose (5.5 mM). Cardiomyocytes from failing and sham hearts were incubated in normoxic conditions with the physiological buffer supplied with 1 mM ouabain to study calcium accumulation during Na+-loading in separate set of experiments. Cell calcium was measured by 45Ca2+ uptake and was related to protein content. Results: Hypoxia-reoxygenation caused 6 fold increase in Ca2+-accumulation in sham cardiomyocytes whereas failing cardiomyocytes did not show any increase. In the presence of 1mM ouabain (normoxic conditions) both sham and failing cardiomyocytes show a similar large increase in calcium accumulation (50-70 fold compared to non-treated cells). Discussion/conclusion: Our results demonstrate a large difference between calcium handling of cardiomyocytes from post MI-failing hearts compared with cardiomyocytes from sham control hearts during hypoxia-reoxygenation. Reoxygenation-mediated calcium uptake is associated with cell death/dysfunction in various cellular and intact heart models of reoxygenation/reperfusion. Interestingly both cell types took up a similar significant amount of calcium in the presence of ouabain (Na+-loading). In conclusion: The present data indicate improved tolerance of the failing cardiomyocytes to hypoxia and reoxygenation. 34 P24 Increased amounts of PP1 and PP2A mediating reduced serine 16 phosphorylation of phospholamban is a possible mechanism for cardiac dysfunction in alveolar hypoxia Karl-Otto Larsen1,2,3, Birgitte Lygren4, Ivar Sjaastad1,3,5, Kurt A. Krobert6, Geir Florholmen1,3, Kristin Arnkværn1,3, Ole Henning Skjønsberg2, Kjetil Taskén4, Geir Christensen1,3 1 2 3 4 5 6 Institute for Experimental Medical Research, Ulleval University Hospital Department of Pulmonary Medicine, Ulleval University Hospital Center for Heart Failure Research, University of Oslo The Biotechnology Centre, University of Oslo Department of Cardiology, Ulleval University Hospital Department of Pharmacology, University of Oslo Introduction: We have previously reported right ventricular (RV) dysfunction and left ventricular (LV) diastolic dysfunction in mice subjected to alveolar hypoxia, and that reduced Serine (Ser) 16 phosphorylated phospholamban (PLB) might be a mechanism for the RV and LV dysfunction. The mechanisms leading to hypophosphorylation of PLB in alveolar hypoxia are not known. Hypothesis: Chronic alveolar hypoxia induces circulating mediators, including cytokines, which lead to changes in the beta-adrenergic pathway or in phosphatases causing reduced Ser16 phosphorylated PLB in both ventricles. Methods: Mice were exposed to 10% oxygen for 2 weeks. Signaling molecules were measured in cardiac tissue, isolated sarcoplasmic reticulum (SR) and serum. Results: Ser16 phosphorylated PLB in the RV and LV free wall was reduced in the hypoxia group to 84 % and 59 % of controls. The levels of beta-adrenergic receptors and adenylyl cyclase activity were unaltered. The amounts of PKA regulatory and catalytic subunits (RI, RII and C), and A kinase anchoring proteins detected by RII-overlay were not changed. However, alveolar hypoxia induced an increase in the amount of PP1 in the RV and the LV free wall to 143% and 177 % of controls. In the LV free wall, PP2A was increased to 124 % of control. The concentrations of IL-18, IL-12p40, RANTES, IL-5, keratinocyte chemoattractant and GM-CSF were increased in serum after 1 day of hypoxia, and IL-18 levels were increased after 2 days, 1 and 2 weeks. Hypoxia increased the amount of Akt, which is involved in IL-18 signaling, to 116 % of control in the RV. Conclusions: Given the unchanged -adrenergic pathway, increased amounts of SR-associated PP1 and PP2A during alveolar hypoxia may be the cause of reduced Ser16 phosphorylated PLB, depressed SR function and cardiac dysfunction. It remains to be determined whether increased IL-18 can influence the observed alterations in phosphatases. 35 Moderated poster session IV: Cardiac and skeletal muscle dysfunction in heart failure P25 Left ventricular systolic torsion by 2-D speckle tracking imaging is load dependent Anders Hodt, Marie Stugaard, Jonny Hisdal, Einar Stranden, Dan Atar, Kjetil Steine P26 Combined treatment with exercise training and losartan ameliorates metabolic dysfunction in the failing heart Morten André Høydal, Ole Johan Kemi, Per Magnus Haram, Anne Garnier, Renee Ventura- Clapier, Øyvind Ellingsen P27 Fatigue in slow twitch skeletal muscle following a dynamic exercise protocol Morten Munkvik, Per Kristian Lunde, Ole M Sejersted P28 Use of single skinned muscle fibres to study the mechanism of increased skeletal muscle fatigability in heart failure Esther Verburg, Morten Munkvik, Per-Kristian Lunde, Graham D. Lamb, Ole M. Sejersted P29 The effect of unilateral knee extensor training on cytokine expression in skeletal muscle of heart failure patients and controls Tommy Aune Rehn, Morten Munkvik, Per Kristian Lunde, Hilde Dishington, Gunnar Slettaløkken, Bjarne Rud, Martin Søkjer, Jostein Hallen, Ole M. Sejersted P30 Calcium handling in skeletal muscle from trained and untrained thigh from heart failure patients and controls Per Kristian Lunde, Tommy Aune Rehn, Morten Munkvik, Almira Karahasan, Gunnar Slettaløkken, Jostein Hallén, Ole M. Sejersted 36 P25 Left ventricular systolic torsion by 2-D speckle tracking imaging is load dependent Anders Hodt1, Marie Stugaard1, Jonny Hisdal2, Einar Stranden2, Dan Atar1, Kjetil Steine1 1 2 Divsion of Cardiology, Aker University Hospital, Oslo Division of Cardiovascular Physiology, Aker University Hospital, Oslo Background: 2-D ultrasound speckle tracking imaging (STI) is a promising tool to assess LV rotation and torsion non-invasively, and correlates well with MRI tagging. The effect of preload on LV torsion is controversial. To explore this issue, we studied 12 healthy men by lower body negative pressure (LBNP). Methods: LBNP was applied using a chamber enclosing lower body to reduce central venous pressure and preload. At baseline and LBNP at -20 and -40 mmHg, LV basal and apical short axes images were acquired to measure LV rotation by STI. LV torsion was defined as difference of LV rotation between apex and base. LBNP at -40 mmHg corresponds to reduction in capillary wedge pressure of about 4 mmHg. Stroke volume (SV) was measured by pulsed Doppler in the LV outflow tract. LV areas were calculated from endocardial traces of short axis images. Results: See table. Stroke volume, ml Heart rate, bpm LV-area end-diastole, cm2 LV-area end-systole, cm2 Peak systolic rotation base, deg Peak systolic rotation apex, deg Peak systolic torsion, deg (Statistical analyses were performed Baseline LBNP -20mmHg 77±20 62±11 52±7 52±7 22.3± 20±3 12±2 11±2 -5.5±3.8 -4.5±3.6 4.8±2.3 4.7±1.7 10.5±3.2 9.8±3.0 by repeated measures ANOVA) LBNP -40mmHg 46±6 57±8 17±3 10±1 -5.7±3.1 3.4±2.4 8.5±3.2 p-value <0.001 <0.01 <0.001 <0.001 0,486 0.086 0.039 Multiple regression showed that the only independent predictor of peak systolic torsion was SV, r = 0.59, r² = 0.35, p < 0.05. The figure shows representative area/torsion loops at baseline (solid line) and LBNP at -40 mmHg (broken line). Conclusion: Peak systolic torsion was significantly reduced, corresponding to a decrease in SV. Reduction of torsion seems to be caused by a decrease in apical rotation. Reduction in preload results in left and downward shift of the area/torsion loop, which indicates that loading is important for LV torsion. 37 P26 Combined Treatment with Exercise Training and Losartan Ameliorates Metabolic Dysfunction in the Failing Heart Morten André Høydal1, Ole Johan Kemi3, Per Magnus Haram1, Anne Garnier4, Renee Ventura- Clapier4, Øyvind Ellingsen1,2 1 2 3 4 Department of Circulation and Medical Imaging, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway Department of Cardiology, St Olavs Hospital, Trondheim, Norway Institute of Biomedical and Life, University of Glasgow, UK. Université Paris Sud, Faculté de Pharmacie, Paris, France Aim: Current clinical approaches for post-myocardial infarction heart failure include exercise training and pharmacological strategies. However, few studies have investigated how combined treatment with those two separates from exercise training or medication alone. Method and results: We established cardiac metabolic dysfunction in postmyocardial infarction heart failure, before starting combined or separate treatment interventions with exercise training and losartan, and Angiotensin II type I (AT1) receptor antagonist. Following left coronary artery ligation in Sprague-Dawley rats, losartan was distributed after 1 week, whereas aerobic interval exercise training at 85-90% of maximal oxygen uptake was initiated 4 weeks post-infarction. Overall, combined treatment yielded best outcome for ventricular pressure characteristics and exercise capacity. Metabolic dysfunction was indicated in myocardial tissue in heart failure, as observed by reduced enzyme activities of creatine and adenylate kinase systems, citrate synthase, cytochrome c oxidase, and, to a lesser degree, lactate dehydrogenase. Gene expression levels of cytochrome c oxidase and peroxisome proliferator-activated receptor gamma coactivator 1 (PGC1), a key regulator of mitochondrial biogenesis and homeostasis tended to decrease in heart failure. Combinatorial treatment with exercise training and losartan partly or fully restored metabolic enzymes and PGC1 gene expression towards healthy levels, whereas exercise training or losartan alone had less order of magnitude effects; in fact, losartan alone appeared to further reduce activity in enzymes involved in energy shuttling and ATP production. Conclusion: Cardiac metabolic dysfunction in heart failure is ameliorated when losartan is given jointly with a controlled, rigorous exercise training program. 38 P27 Fatigue in slow twitch skeletal muscle following a dynamic exercise protocol Morten Munkvik, Per Kristian Lunde, Ole M Sejersted Institute for Experimental Medical Research and Center for Heart Failure Research, University of Oslo, Ullevål University Hospital, N-0407 Oslo, Norway Fatigue mechanisms seem to be different depending on whether the muscle is allowed to shorten (isotonic contractions) or not (isometric contractions) during stimulation. Slow twitch soleus muscles from 12 weeks old male Wistar rats (370±4 g) were stimulated intermittently at 30 Hz (1 sec on 1 sec off for 15 min; 1 ms pulses) in situ at 37ºC and allowed to shorten after the force had reached 1/3 of maximum force (234±4 g). Resting baseline-tension showed a transient rise, reflecting a significant slowing of relaxation that peaked after 100 sec, and returned to near preload values after 5 min. During the first 5 min the shortening declined gradually to about 30 % of the start values. The last ten minutes elapsed with no changes in the recorded parameters. The maximal isometric force-production was reduced after 100 sec, but had regained pre-protocol level after 15 min. Both exercising (EX) and contralateral resting (REST) muscles were frozen and analyzed for metabolites after 100 sec and after 15 min of stimulation. CrP and ATP were significantly lower in EX after 100 sec, but there were no differences compared to REST after 15 min. A “slack test” showed that unloaded shortening velocities were lowest after 100 sec (155 mm/s), and approached pretest values (180 mm/s) after 15 min (165 mm/s), indicating transient changes in the contractile apparatus. Multiplexed proteomics showed that phosphorylation of myosin light chain 2 (MLC2) was reduced at 100sec and remained low. We conclude that isometric muscle dynamics and velocity of unloaded shortening seem to follow CrP and ATP levels, whereas the isotonic fatigue, seen as reduced isotonic shortening, follows the MLC2 phosphorylation level. 39 P28 Use of single skinned muscle fibres to study the mechanism of increased skeletal muscle fatigability in heart failure Esther Verburg1, Morten Munkvik1, Per-Kristian Lunde1, Graham D. Lamb2, Ole M. Sejersted1 1 2 Institute for Experimental Medical Research, Ullevaal University Hospital, Oslo, Norway Dept. of Zoology, La Trobe University, Bundoora Campus, Melbourne, Australia The skinned muscle fibre technique has been used for many years to study the peripheral mechanism of fatigue in healthy skeletal muscle. The technique has contributed to clarifying the roles of for example t-tubuli excitability and lactic acid, ATP, ADP and inorganic phosphate and elevated levels of cytosolic calcium for the development of fatigue. The skinned fibre technique involves the dissection of a section of a single muscle fibre from a whole muscle or a muscle biopsy, followed by the mechanical removal of the outer membrane (sarcolemma) of the muscle cell. In this process the t-tubuli and its connections to the sarcoplasmic reticulum remain intact. The t-tubuli seal off at the sarcolemmal end. The excitation-contraction (EC-) coupling system is fully functional, and the fibers can be stimulated to contract by either depolarisation of the t-tubuli or direct activation of the contractile proteins with Ca2+. Removing the sarcolemma allows free access to the cytosol, and thus the constitution of the cytosol can be manipulated to investigate the roles of acute changes in the concentrations of its components. Altered concentrations of certain ions (for example Mg2+, Na+, K+, Ca2+) or addition of certain components (for example caffeine), can be used to directly stimulate and test certain steps of the EC-coupling system. Tests that can be done include amongst others: [Ca2+]force relationship of the contractile proteins, the capacity of the SR to release and sequester Ca2+, SR Ca2+ content (both total and releasable), force-frequency relationship (electrical stimulation of the t-tubuli), sensitivity of the EC-coupling system to altered [ATP], [ADP], [Pi] and [lactic acid], Ca2+-activated protease (calpain) activity. In our project at the Institute for Experimental Medical Research, we aim to investigate whether the different steps in the EC-coupling system and the mechanism of fatigue in skeletal muscle are altered in heart failure. 40 P29 The effect of unilateral knee extensor training on cytokine expression in skeletal muscle of heart failure patients and controls Tommy Aune Rehn1,2, Morten Munkvik1,2, Per Kristian Lunde1,2, Hilde Dishington1,2, Gunnar Slettaløkken3, Bjarne Rud3, Martin Søkjer4, Jostein Hallen3, Ole M. Sejersted1,2 1 2 3 4 Institute of experimental medical research, Ullevål University Hospital Center for heart failure research Norwegian school of sport sciences Radiological department, Ullevål University Hospital Patients with congestic heart failure (CHF) have increased plasma levels of proinflammatory cytokines that correlate to disease severity and prognosis. Several studies have shown that skeletal muscle can produce cytokines. We hypothesize that skeletal muscle produce inflammatory cytokines to a greater extent in CHF patients compared to controls and that skeletal muscle is a major contributor to the inflammatory status in this patient group. In the present study blood samples and muscle biopsies are taken from CHF patients and controls before and after 6 weeks of unilateral knee extensor training. During a final invasive exercise protocol blood samples from the radial artery and both femoral veins were drawn at rest, during submaximal work of both legs and three minutes after this work to measure arterio-venous cytokine difference. Arteriovenous difference of IL-1, TNF-, IL-18 and IL-6 over trained and untrained leg will be compared with immunohistochemistry for localization- and ELISA on muscle homogenate for quantification of these cytokines. Further, our hypothesis is that endurance training during which the pumping capacity of the heart does not limit muscle blood flow, will attenuate the contribution of muscle to increased circulating levels of proinflammatory cytokines. Preliminary results on IL-18 indicate release from muscle at rest. Interestingly, we see an uptake over untrained leg and release or reduced uptake over the trained leg during exercise. Shortly after cessation of exercise release of IL-18 from the untrained leg greatly exceeded that of the trained leg. It is too early to evaluate possible differences between groups. Further, we hypothesize that endurance training were the oxygen delivery to the working muscle is not limited by the heart’s pumping capacity does not compromise muscle blood flow will attenuate muscle cytokine production. Preliminary results indicate a release of IL-18 from muscle at rest. Interestingly, we see an uptake over untrained leg and a release or lower uptake over trained leg during exercise. Shortly after ended exercise release of IL-18 from untrained leg greatly exceeds that from trained leg. It is too early to evaluate possible differences between groups. 41 P30 Calcium handling in skeletal muscle from trained and untrained thigh from heart failure patients and controls Per Kristian Lunde1, Tommy Aune Rehn1, Morten Munkvik1, Almira Karahasan1, Gunnar Slettaløkken2, Jostein Hallén2, Ole M. Sejersted1 1 2 Institute for Experimental Medical Research, Ullevål University Hospital Norwegian School of Sport Sciences, Oslo, Norway We have previously reported reduced fatigue resistance in slow twitch skeletal muscle from rats with post infarction heart failure (Lunde et al., 2001 and 2002). However, in contrast to normal muscle intracellular calcium release was not related to fatigue development in these rats. We therefore hypothesized that training might affect intracellular calcium cycling differently in muscles from patients with heart failure as compared with controls. Muscle biopsies were taken from the thigh muscle vastus lateralis and analyzed both for content of two isoforms of the calcium pump (SERCA1 and SERCA 2) and the capability of sarcoplasmic reticulum (SR) to take up and release calcium. The subjects then trained one leg on an ergometer for six weeks, and another set of biopsies were taken from vastus lateralis from trained and untrained legs. Endurance of the trained leg became significantly longer compared to the untrained in both heart failure patients and controls. As measured by Western blotting technique the content of the fast calcium pump (SERCA1) was reduced in the trained leg in both groups as compared to the untrained leg. However, the expression of the slow calcium pump of the SR (SERCA2) was significantly increased in the trained leg in both groups. Conclusion: In both heart failure patients and controls endurance training causes a reciprocal shift of the expression of the two isoforms of SERCA towards a slower phenotype. This fits with the prolonged endurance capacity in the trained leg from the subjects. Thus, in heart failure patients endurance training causes a normal shift of the reuptake mechanism for calcium into the SR towards slower rates. References Lunde PK, Dahlstedt AJ, Bruton JD, Lännergren J, Thorén P, Sejersted OM, Westerblad H. Circulation Research 88, 1299-1305. 2001. Lunde PK, Verburg E, Eriksen M, Sejersted OM. Journal of Physiology 540, 571580. 2002. 42 43 List of participants Ademaj Fadil Univ.Hosp.Centre "Mother TEREZA" Afzal Faraz University of Oslo Ahmed M. Shakil University of Oslo Andersen Geir Øystein Ullevaal University Hospital Andersson Kristin B Ullevaal University Hospital Andressen Kjetil Wessel University of Oslo Angel Kristin Aker University Hospital Agewall Stefan Aker University Hospital Ariansen Inger Ullevaal University Hospital Arnkværn Kristin University of Oslo Aronsen Magnus University of Oslo Atar Dan Aker University Hospital Attramadal Håvard University of Oslo Aukrust Pål Rikshospitalet University Hospital Austbø Bjørg University of Oslo Bach Jon Ullevaal University Hospital Bjurstrøm Magne Ski LegeKontor Bjørnerheim Reidar Ullevaal University Hospital Bjørnstad Johannes Ullevaal University Hospital Brattelid Trond University of Oslo Brørs Odd Ullevaal University Hospital Buser Peter University Hospital Basel, Switzerland Carlson Cathrine Ullevaal University Hospital Christensen Geir Ullevaal University Hospital Dahl Mai Britt Ullevaal University Hospital Dahl Christen Peder Rikshospitalet University Hospital Damås Jan Kristian Rikshospitalet University Hospital Dibb Katharine University of Manchester, UK Dickstein Kenneth University of Bergen Dishington Hilde Ullevaal University Hospital Doevendans Pieter Eikemo Hilde University Medical Center, Utrecht, the Netherlands University of Oslo Eisner David University of Manchester, UK Ellingsen Øyvind Estensen Mette-Elise Norwegian University of Science and Technology, Trondheim Rikshospitalet University Hospital Flatebø Torun University of Oslo 44 Florholmen Geir Ullevaal University Hospital Gravning Jørgen University of Oslo Grunnet Morten NeuroSearch A/S Gullestad Lars Rikshospitalet University Hospital Halvorsen Sigrun Ullevaal University Hospital Hansen Rie Schultz NeuroSearch A/S Hasenfuss Gerd University of Göttingen, Germany Haugen Espen Ullevaal University Hospital Hodt Anders Aker University Hospital Hougen Karina Ullevaal University Hospital Husberg Cathrine Ullevaal University Hospital Hussain Rizwan University of Oslo Høydal Morten Ihlen Halfdan Norwegian University of Science and Technology, Trondheim Rikshospitalet University Hospital Iversen Per Ole University of Oslo Jarstadmarken Hilde Ullevaal University Hospital Karahasan Almira Ullevaal University Hospital Knudsen Cathrine Wold Ullevaal University Hospital Koch Walter J. Krobert Kurt Jefferson Medical College, Philadelphia, USA University of Oslo Larsen Karl-Otto Ullevaal University Hospital Levy Finn Olav University of Oslo Louch William E. Ullevaal University Hospital Ludewig Julie University of Oslo Lund Kari Ullevaal University Hospital Lunde Per Kristian Ullevaal University Hospital Mann Douglas Baylor College of Medicine, Houston, USA Martinov Vladimir University of Oslo Moltzau Lise Román University of Oslo Munkvik Morten University of Oslo Mørk Halvor Kjeang Ullevaal University Hospital Nilsson Ullevaal University Hospital Nouri Sharikabad Birgitta Blakstad Mohammad Omland Torbjørn Akershus University Hospital Osnes Jan-Bjørn University of Oslo Qvigstad Eirik University of Oslo Rehn Tommy Aune Ullevaal University Hospital Ullevaal University Hospital 45 Riise Jon University of Oslo Rutkovsky Arkady Ullevaal University Hospital Røsjø Helge Akershus University Hospital Sande Jørn Bodvar University of Oslo Sejersted Ole M. Ullevaal University Hospital Sjåland Cecilie University of Oslo Sjaastad Ivar Ullevaal University Hospital Skomedal Tor University of Oslo Solberg Erik Ekker Diakonhjemmet Hospital Solheim Svein Ullevaal University Hospital Stensløkken Kåre-Olav Ullevaal University Hospital Stokke Mathis Ullevaal University Hospital Strømme Tævje A. Ullevaal University Hospital Swift Fredrik Ullevaal University Hospital Toska Karin University of Oslo Trondsen Roy Ullevaal University Hospital Tønnessen Theis Ullevaal University Hospital Valen Guro University of Oslo Vanberg Paul Aker University Hospital Verburg Esther Ullevaal University Hospital Vistnes Maria Ullevaal University Hospital von Lueder Thomas Aker University Hospital Vaage Jarle Ullevaal University Hospital Westheim Arne Ullevaal University Hospital Winer Lisbeth Ullevaal University Hospital Wæhre Anne University of Oslo Yndestad Arne University of Oslo Ødegaard Annlaug University of Oslo 46