Heart disease is frequently associated with elevated wall

Similar documents
hemodynamic stress. A. Echocardiographic quantification of cardiac dimensions and function in

Regulation of Myocardial Contractility and Cell Size by Distinct PI3K-PTEN Signaling Pathways

Intermittent pressure overload triggers hypertrophy-independent cardiac dysfunction and vascular rarefaction

Journal of the American College of Cardiology Vol. 45, No. 11, by the American College of Cardiology Foundation ISSN /05/$30.

Fetal gene upregulation by 1-wk TAC is significantly increased in mice lacking RGS2.

C57BL/6 Mice are More Appropriate. than BALB/C Mice in Inducing Dilated Cardiomyopathy with Short-Term Doxorubicin Treatment

c Ischemia (30 min) Reperfusion (8 w) Supplementary Figure bp 300 bp Ischemia (30 min) Reperfusion (4 h) Dox 20 mg/kg i.p.

In Vivo Animal Models of Heart Disease. Why Animal Models of Disease? Timothy A Hacker, PhD Department of Medicine University of Wisconsin-Madison

Supplemental Figure I

J Jpn Coll Angiol, 2009, 49:

Adenylyl cyclase (AC) is the effector molecule in

2013 W. H. Freeman and Company. 12 Signal Transduction

Supplement Material. chain (MLC2v) promoter to generate cardiac-specific MKK4 knockout mice (referred to

The phosphoinositide 3-kinase (PI3K) system has a fundamental

SUPPLEMENTAL MATERIAL. Supplementary Methods

Gallic acid prevents isoproterenol-induced cardiac hypertrophy and fibrosis through regulation of JNK2 signaling and Smad3 binding activity

Supplementary Figure 1. Confocal immunofluorescence showing mitochondrial translocation of Drp1. Cardiomyocytes treated with H 2 O 2 were prestained

Postn MCM Smad2 fl/fl Postn MCM Smad3 fl/fl Postn MCM Smad2/3 fl/fl. Postn MCM. Tgfbr1/2 fl/fl TAC

LV geometric and functional changes in VHD: How to assess? Mi-Seung Shin M.D., Ph.D. Gachon University Gil Hospital

Heart Failure (HF) Treatment

Tcf21 MCM ; R26 mtmg Sham GFP Col 1/3 TAC 8W TAC 2W. Postn MCM ; R26 mtmg Sham GFP Col 1/3 TAC 8W TAC 2W

Qualifying Examination (Part I)

(Stratagene, La Jolla, CA) (Supplemental Fig. 1A). A 5.4-kb EcoRI fragment

BNP mrna expression in DR and DS rat left ventricles (n = 5). (C) Plasma norepinephrine

AT1 RECEPTOR BLOCKADE ATTENUATES INSULIN RESISTANCE AND MYOCARDIAL REMODELING IN RATS WITH DIET-INDUCED OBESITY

1. Cardiomyocytes and nonmyocyte. 2. Extracellular Matrix 3. Vessels שאלה 1. Pathobiology of Heart Failure Molecular and Cellular Mechanism

SUPPLEMENTARY INFORMATION

Biosignals, Chapter 8, rearranged, Part I

Signal Transduction Cascades

Supplementary Figures Supplementary Figure 1. Development of the camp biosensor targeted to the SERCA2a microdomain.

Genetic ablation of Acp1 (Lmptp) in mice prevents heart failure

Index. A Action potential duration, increased, by decreases in sodium current,

Sphingosine-1-phosphate signaling and cardiac fibrosis. Department of Physiology, Kanazawa University School of Medicine, Kanazawa, Japan

SUPPLEMENTAL DATA. Lumen area ( m 2 )

Signal Transduction: G-Protein Coupled Receptors

שינויים מולקולאריים ומבניים באי ספיקת לב אפשרויות לטיפול עתידני

Drug Receptor Interactions and Pharmacodynamics

Accelerated Cardiomyopathy in Mice With Overexpression of Cardiac G s and a Missense Mutation in the -Myosin Heavy Chain

Exercise in Adverse Cardiac Remodeling: of Mice and Men

Uncovering the mechanisms of wound healing and fibrosis

Remodeling the failing heart: : the biology and future treatment options

MEK-ERK pathway modulation ameliorates disease phenotypes in a mouse model of Noonan syndrome associated with the Raf1 L613V mutation

Philipp Schlegel, Jan Ksienzyk, Jens Barthelmes, Uwe Haberkorn, Walter J. Koch, Hugo A. Katus, Patrick Most, Oliver J. Mueller, Philip W.J.

Protection against doxorubicin-induced myocardial dysfunction in mice by cardiac-specific expression of carboxyl terminus of hsp70-interacting protein

In the name of GOD. Animal models of cardiovascular diseases: myocardial infarction & hypertension

What would you observe if you fused a G1 cell with a S cell? A. Mitotic and pulverized chromosomes. B. Mitotic and compact G1 chromosomes.

Ca 2+ - and mitochondrial-dependent cardiomyocyte necrosis as a primary mediator of heart failure

Myocardial lipid accumulation and lipotoxicity in heart failure

Supplemental Figure 1. Western blot analysis indicated that MIF was detected in the fractions of

G-Protein Coupled Receptors GPCRs. GPCRs

Probe. Hind III Q,!&#12?R'!! /0!!!!D1"?R'! vector. Homologous recombination

Supporting Information

A particular set of insults induces apoptosis (part 1), which, if inhibited, can switch to autophagy. At least in some cellular settings, autophagy se

The -adrenergic receptor ( AR) signaling pathway is one of

Original Article Protective effect of ginsenoside Rg1 on isoproterenol-induced acute myocardial ischemia in rats

Qualifying Examination (Part I)

RT-100 technical summary (as of March 2015) Table of contents

Lecture 15. Signal Transduction Pathways - Introduction

Lecture: CHAPTER 13 Signal Transduction Pathways

Kidney. Heart. Lung. Sirt1. Gapdh. Mouse IgG DAPI. Rabbit IgG DAPI

ß-Adrenergic Receptor Signaling in Congestive Heart Failure. Emily Baxter. Professor Srikant. McGill University EXMD 401.

Protein kinase A mediated stimulation of activating transcription factor 3 by hypertrophic stimuli in cardiomyocytes

Full Record.

Growth and Differentiation Phosphorylation Sampler Kit

Mechanisms of heart failure with normal EF Arterial stiffness and ventricular-arterial coupling. What is the pathophysiology at presentation?

Supplementary Figure 1. Spatial distribution of LRP5 and β-catenin in intact cardiomyocytes. (a) and (b) Immunofluorescence staining of endogenous

Research article. Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Health Sciences University, Augusta, Georgia, USA.

Hormones and Signal Transduction. Dr. Kevin Ahern

Myocardial Infarction

Large myocardial infarction (MI) is a major cause of

Vets 111/Biov 111 Cell Signalling-2. Secondary messengers the cyclic AMP intracellular signalling system

G-Protein Signaling. Introduction to intracellular signaling. Dr. SARRAY Sameh, Ph.D

Cardiac Gene Therapy: Beyond the Mouse. David M Kaye Heart Failure Research Group Baker IDI, Melbourne, AUSTRALIA

Myocardial fibrosis is a pathological feature associated

renoprotection therapy goals 208, 209

FOCUS ON CARDIOVASCULAR DISEASE

Left ventricular (LV) remodeling after myocardial infarction

Targeted inhibition of p38 MAPK promotes hypertrophic cardiomyopathy through upregulation of calcineurin-nfat signaling

Model Answer. M.Sc. Zoology (First Semester) Examination Paper LZT 103 (Endocrinology)

Cell Communication. Cell Communication. Communication between cells requires: ligand: the signaling molecule

Value of echocardiography in chronic dyspnea

Cell Signaling part 2

Drugs Used in Heart Failure. Assistant Prof. Dr. Najlaa Saadi PhD pharmacology Faculty of Pharmacy University of Philadelphia

Propagation of the Signal

International Graduate Research Programme in Cardiovascular Science

(A) PCR primers (arrows) designed to distinguish wild type (P1+P2), targeted (P1+P2) and excised (P1+P3)14-

INTERACTION DRUG BODY

Small Platform Catheter-Based Left Ventricular Assist Device Support Suppresses Cardioprotective Beta Arrestin- Mediated Signal Transduction

Serum cytokine levels in control and tumor-bearing male and female mice at day 15.

E10.5 E18.5 P2 10w 83w NF1 HF1. Sham ISO. Bmi1. H3K9me3. Lung weight (g)

Epac1-dependent phospholamban phosphorylation mediates the cardiac response to stresses

SUPPLEMENTARY INFORMATION

β-adrenergic receptor blockade arrests myocyte damage and preserves cardiac function in the transgenic G sα mouse

Echo assessment of the failing heart

Receptor mediated Signal Transduction

The PI3K/AKT axis. Dr. Lucio Crinò Medical Oncology Division Azienda Ospedaliera-Perugia. Introduction

The cyclic GMP (cgmp) signaling pathway is a ubiquitous

Membrane Structure and Function

The elements of G protein-coupled receptor systems

Atrogin-1/muscle atrophy F-box inhibits calcineurin-dependent cardiac hypertrophy by participating in an SCF ubiquitin ligase complex

Transcription:

Phosphoinositide 3-Kinase Deficient Mice Are Protected From Isoproterenol-Induced Heart Failure Gavin Y. Oudit, MSc, MD; Michael A. Crackower, PhD; Urs Eriksson, MD; Renu Sarao, MSc; Ivona Kozieradzki, MSc; Takehiko Sasaki, MD; Junko Irie-Sasaki, MD; Dominica Gidrewicz, MSc; Vitalyi O. Rybin, PhD; Teiji Wada, MD; Susan F. Steinberg, MD; Peter H. Backx, DVM, PhD; Josef M. Penninger, MD Background We have recently shown that genetic inactivation of phosphoinositide 3-kinase (PI3K ), the isoform linked to G-protein coupled receptors, results in increased cardiac contractility with no effect on basal cell size. Signaling via the G-protein coupled -adrenergic receptors has been implicated in cardiac hypertrophy and heart failure, suggesting that PI3K might play a role in the pathogenesis of heart disease. Methods and Results To determine the role for PI3K in hypertrophy induced by G-protein coupled receptors and cardiomyopathy, we infused isoproterenol, a -adrenergic receptor agonist, into PI3K -deficient mice. Compared with controls, isoproterenol infusion in PI3K -deficient mice resulted in an attenuated cardiac hypertrophic response and markedly reduced interstitial fibrosis. Intriguingly, chronic -adrenergic receptor stimulation triggered impaired heart functions in wild-type mice, whereas PI3K -deficient mice retained their increased heart function and did not develop heart failure. The lack of PI3K attenuated the activation of Akt/protein kinase B and extracellular signal-regulated kinase 1/2 signaling pathways in cardiac myocytes in response to isoproterenol. 1 - and 2 -adrenergic receptor densities were decreased by similar amounts in PI3K -deficient and control mice, suggesting that PI3K isoform plays no role in the downregulation of -adrenergic receptors after chronic -adrenergic stimulation. Conclusions Our data show that PI3K is critical for the induction of hypertrophy, fibrosis, and cardiac dysfunction function in response to -adrenergic receptor stimulation in vivo. Thus, PI3K may represent a novel therapeutic target for the treatment of decreased cardiac function in heart failure. (Circulation. 2003;108:2147-2152.) Key Words: heart failure signal transduction hypertrophy Heart disease is frequently associated with elevated wall stress as a result of pressure overload, such as in hypertension or volume overload seen in valvular disorders. Increased wall stress in the heart triggers a hypertrophic response. 1 What initially reflects a compensatory response turns into pathological hypertrophy, which eventually leads to cardiac decompensation and heart failure. The signaling molecules that regulate the hypertrophic response and cardiac function under pathologic conditions are not well understood. 1 In flies and mammals, hypertrophic responses can be initiated via phosphoinositide 3-kinase (PI3K) signaling pathways. 2,3 PI3Ks constitute a family of evolutionarily conserved lipid kinases that regulate a vast array of fundamental cellular responses, including proliferation, adhesion, cell size, and protection from apoptosis. 4 Activated PI3K phosphorylates the 3 -position of phosphatidylinositol (PtdIns)(4,5)P 2, leading to the generation of PtdIns(3,4,5)P 3, which activates multiple downstream targets, including Akt/protein kinase B (PKB). 4 6 Four different type I PI3Ks have been described, 3 of which (PI3K,, and ) (class I A ) are activated by receptor tyrosine kinase pathways. In contrast, PI3K is activated by the subunit of G-proteins and acts downstream of G- protein coupled receptors (GPCRs) (class I B ). 3,4,7 The tumor suppressor PTEN (phosphatase and tensin homologue on chromosome ten) is a lipid phosphatase that dephosphorylates the D3 position of PtdIns(3,4,5)P 3 and therefore antagonizes the activity of the PI3K isoforms in a wide range of cell types, including cardiomyocytes. 3,8 Recent genetic evidence indicates that PI3K is the sole PI3K that couples to GPCRs and its function can be uncoupled from the class I A PI3K isoforms. 3,5,6 We have recently Received December 12, 2002; de novo received April 28, 2003; revision received June 3, 2003; accepted June 17, 2003. From the Departments of Physiology and Medicine (G.Y.O., D.G., P.H.B.) and Medical Biophysics and Immunology (U.E., R.S., I.K., T.W., J.M.P.), University Health Network, University of Toronto, Ontario, Canada; Department of Functional Genomics (M.A.C.), Amgen Inc, Thousand Oaks, Calif; Institute of Molecular Biotechnology of the Austrian Academy of Sciences (U.E., R.S., I.K., T.W., J.M.P.), Vienna, Austria; Department of Pharmacology (T.S., J.-I.S.), The Tokyo Metropolitan Institute of Medical Science, Japan; PRESTO (T.S., J.-I.S.), Japan Science and Technology Corporation (T.S., J.-I.S.), Tokyo, Japan; and Department of Pharmacology (V.O.R., S.F.S.), College of Physicians and Surgeons, Columbia University, New York, NY. Correspondence to J.M. Penninger, University Health Network, University of Toronto, 620 University Ave, Toronto, Ontario M5G 2C1, Canada. E-mail jpenning@uhnres.utoronto.ca 2003 American Heart Association, Inc. Circulation is available at http://www.circulationaha.org DOI: 10.1161/01.CIR.0000091403.62293.2B 2147

2148 Circulation October 28, 2003 reported that mice deficient for p110, the catalytic subunit of PI3K, display increased cardiac contractility but normal heart size via increased camp signaling. 3 Moreover, we showed that cardiac hypertrophy and myocardial contractility can be genetically uncoupled. 3 However, sustained adrenergic stimulation has been implicated in the development of pathological hypertrophy and progression of heart failure in animal models 7,9,10 and humans, 10 12 suggesting that PI3K may play a role in agonist-induced cardiac hypertrophy and disease. Increased cardiac contractility seen in p110 / hearts also suggests that loss of PI3K function may protect from GPCR-induced heart failure. In the present study, we investigated the cardiac responses of the PI3K -deficient mice to chronic -adrenergic stimulation. Methods Mice p110 / mice have previously been described. 5 Hypertrophy and heart failure were induced by chronic infusion of isoproterenol (Sigma) for 7 days at a dose of 15 mg/kg per d using Alzet osmotic mini-pumps. Pumps were removed 24 hours before echocardiography and hemodynamic measurements. All experiments were performed in accordance with institutional guidelines. Only littermate male mice of 10 to 12 weeks of age were used. Heart Morphometry, TUNEL Assay, Echocardiography, and Hemodynamic Measurements For heart morphometry, hearts were arrested with KCl, fixed with 10% buffered formalin, and embedded in paraffin. Myocardial interstitial fibrosis was determined as collagen volume fraction (PSR-stained sections) using color-subtractive computer-assisted image analysis (Image Processing Tool Kit version 2.5). In situ DNA fragmentation was labeled using the TUNEL assay (ApopTag Plus kit) (Oncor). After removal of the osmotic pumps for 24 hours, echocardiographic assessments and invasive hemodynamic measurements were carried out as previously described. 3 Signaling, mrna, and Western Blot Analyses Total RNA was prepared from hearts using Trizol, resolved on a 0.8% formamide gel, blotted to nylon membranes (Amersham), and Figure 1. Signaling and cardiac hypertrophy responses in p110 / and p110 / mice. A, Heart sections from p110 / and p110 / mice treated with vehicle or isoproterenol for 7 days. B, Quantitation of heart to body weight ratio and heart weight to tibial length ratio in vehicle and isoproterenol-treated mice; n 9 per group. *P 0.05; **P 0.01 compared with vehicle. C, Northern blot analysis and quantitation of cardiac hypertrophy markers in p110 / and p110 / mice treated with vehicle (1) or isoproterenol (2) for 7 days. GAPDH expression was used as a loading control. Baseline levels of the indicated markers were comparable between p110 / and p110 / hearts. D, ERK1/2 phosphorylation in isolated p110 / and p110 / neonatal cardiomyocytes. Cells were (1) left untreated or stimulated with (2) isoproterenol (10 mol/l) and (3) bfgf (250 ng/ml) for 10 minutes. Western blot data are shown. E, Impaired Akt/PKB activation in total heart extracts from p110 / and p110 / mice treated with vehicle or isoproterenol for 7 days; n 5 per group. *P 0.01 between all groups. Values are mean SEM. MyHC indicates myosin heavy chain; ANF, atrial natriuretic factor; and BNP, brain natriuretic peptide.

Oudit et al PI3K and -Adrenergic Signaling 2149 Echocardiographic and Hemodynamic Parameters in Littermate p110 / and p110 / Mice After Chronic -Adrenergic Receptor Stimulation p110 / Vehicle p110 / Vehicle p110 / Isop p110 / Isop HR, bpm 521 13.5 503 14.8 491 17.2 513 9.3 AW, mm 0.693 0.019 0.696 0.015 0.879 0.02* 0.733 0.017 PW, mm 0.687 0.016 0.706 0.022 0.884 0.021* 0.742 0.018 LVEDD, mm 3.98 0.11 4.03 0.12 4.15 0.08 3.96 0.1 LVESD, mm 2.16 0.09 1.73 0.06 2.89 0.07* 1.95 0.09 LVM/BW, mg/g 3.16 0.18 3.2 0.17 4.79 0.13* 3.65 0.11 FS, % 46.7 1.2 57.8 1.3 29.6 1.9* 50.3 1.4 PAV, cm/s 87.5 3.2 98.9 5.1 68.6 3.4* 92.5 2.9 VCFc, circ/s 8.26 0.27 11.3 0.42 5.43 0.31* 10.3 0.38 SBP, mm Hg 108.3 2.9 112 3.6 75.6 3.3* 107.8 3.2 MABP, mm Hg 83.4 3 85.4 2.2 53.2 2.4* 81.4 2.9 LVEDP, mm Hg 5.84 0.3 5.65 0.44 10.9 0.86* 6.55 0.58 dp/dt max, mm Hg/sec 5995 120 8984 241 3142 294* 7963 257 dp/dt min, mm Hg/sec 5487 128 8093 257 2706 218* 6987 216 Isop indicates isoproterenol; HR, heart rate; AW, anterior left ventricular wall thickness; PW, posterior left ventricular wall thickness; LVEDD, left ventricular end diastolic diameter; LVESD, left ventricular end systolic diameter; LVM, left ventricular mass; BW, body weight; FS, fractional shortening; PAV, peak aortic velocity; VCFc, velocity of circumferential shortening corrected for heart rate; SBP, systolic blood pressure; MABP, mean arterial blood pressure; LVEDP, left ventricular end-diastolic pressure; dp/dt max, maximum first derivative of the change in left ventricular pressure; and dp/dt min, minimum first derivative of the change in left ventricular pressure. n 8 for all groups; values are mean SEM. *P 0.01 compared with all other groups; P 0.01 compared with p110 / vehicle group; P 0.05 compared with p110 / vehicle group. probed with cdna probes for atrial natriuretic factor, brain natriuretic peptide, -myosin heavy chain, and GAPDH. Akt/PKB kinase activity was measured using a commercial in vitro kinase assay kit according to the manufacturer s protocol (Upstate Biotechnologies). 5 Ventricular cardiomyocytes were isolated as described. 3 Experiments were carried out 48 to 72 hours after plating and removal of fibroblasts. Extracellular signal-regulated kinase (ERK) 1/2 phosphorylation and ERK1/2 protein levels were determined by Western blotting (Cell Signaling Technology). -Adrenergic Receptor Densities Mice were anesthetized with isoflurane and O 2, hearts were removed, and ventricular tissue was excised and frozen. Samples were processed, and light sarcolemmae were not treated ( ) or treated with peptide N-glycosidase F (PNG-F) ( ) and subjected to SDS-PAGE as previously described. 13 Protein recovery was similar for p110 / and p110 / ventricles without or with isoproterenol treatment, and equal amounts of protein loading were used for each group. In addition, all samples were run at 2 (for 1 -adrenergic) and 3 (for 2 -adrenergic) different amounts of protein loading in a single experiment to avoid interassay variability. Statistical Analysis One-way ANOVA was used to test for overall significance, followed by the Student Newman-Keuls test for multiple comparison testing between the various groups. All statistical analyses were performed with the SPSS software (version 10.1). Data are presented as mean SEM; n refers to the sample size. Results Loss of p110 Impairs the Hypertrophic Response to Isoproterenol To determine the role of PI3K in GPCR-induced cardiac hypertrophy and cardiomyopathy, we chronically infused littermate p110 / (knockout) and p110g / (control) mice with the -adrenergic receptor agonist isoproterenol. Control mice displayed a marked increase in heart size (Figure 1A), heart to body weight ratio, heart to tibial length ratio (Figure 1B), and left ventricular mass and wall thickness (Table). In isoproterenol-infused p110 / mice, the hypertrophic response still occurred but was markedly attenuated (Figures 1A and 1B) (Table). Importantly, measurements of cardiomyocyte sizes in cross-sections confirmed impaired cardiomyocyte hypertrophy in p110 / mice (Figure 2D). Cardiac hypertrophy is associated with prototypical alterations in gene expression. 1 We therefore assessed the mrna levels of hypertrophy markers in hearts of p110 / and p110 / mice chronically stimulated with isoproterenol. Intriguingly, despite the attenuation of hypertrophy in p110 / hearts, we observed a similar alteration in expression profiles in atrial natriuretic factor, brain natriuretic peptide, and -myosin heavy chain compared with p110 / hearts (Figure 1C). It is unclear whether PI3K is essential for GPCR signaling in cardiomyocytes. 7,14 We therefore stimulated purified ventricular cardiomyocytes from p110 / and p110 / littermates with the GPCR-agonist isoproterenol or the tyrosine kinase based receptor agonist basic fibroblast growth factor (bfgf) (Figure 1D). Whereas bfgf-induced ERK1/2 activation occurred normally, isoproterenol-induced ERK1/2 phosphorylation was completely abolished in p110 / cardiomyocytes (Figure 1D). Similarly, in p110 / hearts, chronic isoproterenol infusion did not increase endogenous Akt/PKB activity (Figure 1E).

2150 Circulation October 28, 2003 Figure 2. Loss of p110 protects from interstitial fibrosis and hypertrophy. A, Heart sections from p110 / and p110 / mice treated with vehicle or isoproterenol (ISO) for 7 days. Trichrome and picrosirius red (PSR) staining to detect remodeling and interstitial fibrosis are shown. B, Morphometric quantitation of fibrosis in vehicle- and isoproterenol-treated mice. n 5 per group. *P 0.01 compared with vehicle. C, Quantitation of apoptosis in vehicle- and isoproterenol-treated mice. n 3 per group, with 3 sections from each heart. *P 0.05 compared with vehicle. D, Quantitation of cardiomyocyte size based on cross-sectional area in vehicle- and isoproterenoltreated mice. n 3 per group, with 3 sections from each heart. *P 0.05; **P 0.01 compared with vehicle. Values are mean SEM. Isoproterenol Stimulation Leads to Reduced Interstitial Fibrosis in p110 -Deficient Mice Decompensation of the hypertrophic myocardium and the progression to cardiomyopathy is accompanied by increased fibrosis and disorganization of myocytes. 1 We therefore carried out histological analysis to study the cardiac remodeling response. In control hearts (p110 / ), treatment with isoproterenol resulted in disorganization of the myocytes and a marked increase in cardiac fibrosis (Figure 2A). Quantification of interstitial fibrosis showed a significant increase in collagen deposition in the hearts of isoproterenol-treated control mice (Figure 2B). By contrast, p110 / mice displayed no significant increase in cardiac fibrosis or alteration in myocyte organization (Figures 2A and 2B). Cardiomyocyte cell death was comparable between isoproterenol-treated p110 / and p110 / mice (Figure 2C). Isoproterenol-Induced Cardiovascular Dysfunction Is Reduced in p110 -Deficient Mice Given the marked differences in signaling and hypertrophic and fibrotic responses, we studied the functional changes in heart function in mice infused with isoproterenol after withdrawal of the drug. Chronic infusion of isoproterenol into p110 / mice led to a significant reduction in heart function with decreased fractional shortening, velocity of circumferential fiber shortening, and peak aortic velocity (Table). Invasive hemodynamic measurements showed increased left ventricular end-diastolic pressure and decreases in dp/dt max and blood pressure in p110 / animals (Table), again indicating severe impairment of heart function. By contrast, chronic isoproterenol stimulation of p110 / mice caused a markedly smaller reduction in cardiac function and blood pressures. Moreover, these hearts retained an increased pump function relative to untreated control hearts (Table).

Oudit et al PI3K and -Adrenergic Signaling 2151 Figure 3. Ventricular -adrenergic receptor densities. A, Immunoblots on purified sarcolemmal preparations from p110 / and p110 / mice treated with vehicle or isoproterenol for 7 days. Purified sarcolemmal preparations were enriched in the specific markers, such as Na-K ATPase, as well as caveolin-1 and caveolin-3 (not shown). Immunoblot analysis was with anti-adenylyl cyclase (AC) V/VI (top, 20 g protein loaded), anti 1 -adrenergic (middle, 20 g protein loaded), or anti 2 -adrenergic (bottom, 100 g protein loaded) antibodies or after antibody preblocking with antigen ( Antigen). Vehicle-treated samples are designated as control. The antibody used for the AC cannot discriminate between ACV and ACVI isoforms. Equal amounts of protein loading were used for each group, and the relative changes in -adrenergic densities were independent of the amount of protein loading. B, Quantitations of ACV, ACVI, 1 -adrenergic ( 1 -AR), and 2 -adrenergic ( 2 -AR) receptor expression (n 6 per group). Western blots were scanned from film with a laser densitometer (Molecular Dynamics) and quantified using ImageQuant software (Molecular Dynamics). Values are mean SEM. *P 0.05. Lack of Differential Downregulation of -Adrenergic Receptors in p110 -Deficient Mice PI3K signaling has been proposed to mediate -adrenergic downregulation after agonist stimulation. 7,14 1 - and 2 - adrenergic receptors were downregulated by 75% and 50%, respectively, in membrane fractions from p110 / hearts (ventricular tissue) from mice infused with isoproterenol compared with vehicle-treated mice (Figures 3A and 3B). Importantly, there were no differential changes in 1 -or 2 -adrenergic receptor expression in membrane fractions from p110 / and p110 / hearts (ventricular tissue) at baseline and in response to chronic in vivo isoproterenol stimulation (Figures 3A and 3B). Expression levels of the major cardiac adenylate cyclase isoforms V and VI were also comparable between p110 / and p110 / hearts and were not affected by chronic -adrenergic activation (Figures 3A and 3B). Discussion Excessive catecholamines and adrenergic stimulation have been clearly linked to cardiac hypertrophy and disease. Indeed, altered signaling via the G-protein coupled -adrenergic receptors has been implicated in heart failure in animal models 7,9,10 and humans. 10 12 Although the increased protein expression of PTEN in response to chronic isoproterenol stimulation may serve to negatively regulate the activity of PI3K, 3,15 the precise role of PI3K isoform in heart failure remains unresolved. We have shown previously that loss of p110 leads to an increase in cardiomyocyte contractility. 3 Our present data show that PI3K, the PI3K-isoform linked to GPCRs, is critical for the induction of myocardial hypertrophy, interstitial fibrosis, and cardiac dysfunction in response to -adrenergic receptor stimulation in vivo. Importantly, p110 / mice display a resistance to the effects of isoproterenol on cardiac structure and function. Because it has been suggested that an increase in cardiac function may serve to improve myocardial performance in models of dilated cardiomyopathy, 1,7 we confirmed that p110 / hearts maintained their hypercontractility after chronic adrenergic stimulation. However, the induction of several hypertrophy markers in response to isoproterenol was not affected by the loss of p110, which suggests that p110 is not required for alteration in fetal gene expression seen in myocardial hypertrophy and that these genetic changes can be uncoupled from the hypertrophic process. Our data show that chronic activation of -adrenergic signaling results in markedly decreased heart function and the onset of cardiomyopathy in p110 / mice. In contrast, loss of p110 renders animals resistant to increased interstitial fibrosis and cardiac dysfunction. Akt/protein kinase B (PKB), a major molecular target of the PI3K signaling pathway, is involved in cell hypertrophy in multiple systems, 2,3,16 and the

2152 Circulation October 28, 2003 mitogen-activated protein kinase ERK1/2 also plays a central role in cardiac hypertrophy. 17 Genetic evidence in hematopoetic cells shows that PI3K is the sole PI3K that couples to GPCRs. 5,6 Importantly, infusion of isoproterenol into p110 / mice showed that -adrenergic receptor stimulation induced in vivo activation of Akt/PKB and treatment of isolated cardiomyocytes with isoproterenol leads to activation of ERK1/2. These genetic data demonstrate for the first time that after GPCR stimulation, PI3K is the essential PI3K isoform required for Akt/PKB and ERK1/2 activation in cardiomyocytes, which is consistent with our data in hematopoietic cells. 5 In other models of heart disease (pressure-overload hypertrophy), there is also activation of Akt/PKB via the PI3K isoform. 18 As such, whereas the PI3K isoform plays no role in the control of basal (physiological growth) Akt/PKB levels, 3 under GPCR stimulation, there is clearly activation of Akt/PKB via PI3K, possibly via the interaction with G subunits. 19 Consistent with our recent observations, the intact stimulation of ERK1/2 by the tyrosine receptor agonist bfgf in p110 / cardiomyocytes confirms that the activity of PI3K class I A isoforms (PI3K,, and ) can be genetically uncoupled from the class I B isoform, PI3K. 3 Despite the lack of activation of Akt/PKB and ERK1/2 pathways, which are clearly antiapoptotic pathways, 4,17 apoptosis was not increased in the p110 / mice, which is likely attributable to the antiapoptotic effects mediated by enhanced 2 -adrenergic receptor signaling in p110 / cardiomyocytes. 3,7 Because PI3K also has a role in GPCR signaling in isolated cardiac fibroblasts, 20 it is possible that the reduced interstitial fibrosis in the heart is the direct consequence of attenuated -adrenergic receptor mediated stimulation of cardiac fibroblasts in the p110 / hearts. Recently published reports suggest that PI3K might be involved in -adrenergic receptor downregulation (internalization). 14,18 However, the loss of PI3K has no apparent effect on the downregulation of -adrenergic receptors in response to chronic adrenergic stimulation. However, we cannot rule out the role of other PI3K isoforms in the downregulation of -adrenergic receptors, and there exists the distinct possibility that receptor desensitization could be differentially affected, ie, functional responses are different. 7,14 In summary, we have shown that the PI3K has an important role to maintain heart function under the pathologic condition of chronic adrenergic stimulation, and our results represent the first genetic evidence for a potential therapeutic role of the modulation of PI3K signaling in heart failure. The role of PI3K inhibition in other genetic and acquired models of heart failure and the mechanisms of cardiac protection such as the role of PI3K in intracellular Ca 2 regulation (eg, phosphorylation state of phospholamban) 1,3 and the reduction in interstitial fibrosis need be further evaluated. Acknowledgments This work was supported by the Institute for Molecular Biotechnology of the Austrian Academy of Sciences, the Heart and Stroke Foundation of Canada, and the Canadian Institute for Health Research. G.Y. Oudit is a recipient of a postdoctoral fellowship from the Canadian Institute for Health Research and the Heart and Stroke Foundation of Canada. References 1. Hunter JJ, Chien KR. Signaling pathways for cardiac hypertrophy and failure. N Engl J Med. 1999;341:1276 1283. 2. Shioi T, Kang PM, Douglas PS, et al. The conserved phosphoinositide 3-kinase pathway determines heart size in mice. EMBO J. 2000;19: 2537 2548. 3. Crackower MA, Oudit GY, Kozieradzki I, et al. Regulation of myocardial contractility and cell size by distinct PI3K-PTEN signaling pathways. Cell. 2002;110:737 749. 4. Toker A, Cantley LC. Signalling through the lipid products of phosphoinositide-3-oh kinase. Nature. 1997;387:673 676. 5. Sasaki T, Irie-Sasaki J, Jones RG, et al. Function of PI3Kgamma in thymocyte development, T cell activation, and neutrophil migration. Science. 2000;287:1040 1046. 6. Hirsch E, Katanaev VL, Garlanda C, et al. Central role for G proteincoupled phosphoinositide 3-kinase gamma in inflammation. Science. 2000;287:1049 1053. 7. Rockman HA, Koch WJ, Lefkowitz RJ. Seven-transmembrane-spanning receptors and heart function. Nature. 2002;415:206 212. 8. Maehama T, Dixon JE. The tumor suppressor, PTEN/MMAC1, dephosphorylates the lipid second messenger, phosphatidylinositol 3,4,5- trisphosphate. J Biol Chem. 1998;273:13375 13378. 9. Scheuer J. Catecholamines in cardiac hypertrophy. Am J Cardiol. 1999; 83:70H 74H. 10. Brede M, Wiesmann F, Jahns R, et al. Feedback inhibition of catecholamine release by two different alpha2-adrenoceptor subtypes prevents progression of heart failure. Circulation. 2002;106:2491 2496. 11. Small KM, Wagoner LE, Levin AM, et al. Synergistic polymorphisms of beta1- and alpha2c-adrenergic receptors and the risk of congestive heart failure. N Engl J Med. 2002;347:1135 1142. 12. Hajjar RJ, MacRae CA. Adrenergic-receptor polymorphisms and heart failure. N Engl J Med. 2002;347:1196 1199. 13. Rybin VO, Xu X, Lisanti MP, et al. Differential targeting of beta -adrenergic receptor subtypes and adenylyl cyclase to cardiomyocyte caveolae: a mechanism to functionally regulate the camp signaling pathway. J Biol Chem. 2000;275:41447 41457. 14. Naga Prasad SV, Laporte SA, Chamberlain D, et al. Phosphoinositide 3-kinase regulates beta2-adrenergic receptor endocytosis by AP-2 recruitment to the receptor/beta-arrestin complex. J Cell Biol. 2002;158: 563 575. 15. Schwartzbauer G, Robbins J. The tumor suppressor gene PTEN can regulate cardiac hypertrophy and survival. J Biol Chem. 2001;276: 35786 35793. 16. Shioi T, McMullen JR, Kang PM, et al. Akt/protein kinase B promotes organ growth in transgenic mice. Mol Cell Biol. 2002;22:2799 2809. 17. Bueno OF, Molkentin JD. Involvement of extracellular signal-regulated kinases 1/2 in cardiac hypertrophy and cell death. Circ Res. 2002;91: 776 781. 18. Naga Prasad SV, Esposito G, Mao L, et al. G -dependent phosphoinositide 3-kinase activation in hearts with in vivo pressure overload hypertrophy. J Biol Chem. 2000;275:4693 4698. 19. Bommakanti RK, Vinayak S, Simonds WF. Dual regulation of Akt/ protein kinase B by heterotrimeric G protein subunits. J Biol Chem. 2000;275:38870 38876. 20. Kim J, Eckhart AD, Eguchi S, et al. Beta-adrenergic receptor-mediated DNA synthesis in cardiac fibroblasts is dependent on transactivation of the epidermal growth factor receptor and subsequent activation of extracellular signal-regulated kinases. J Biol Chem. 2002;277:32116 32123.