Mitral Regurgitation Augments Post-Myocardial …Methods Apical MIs were created in 12 sheep, and 6...

11
PRECLINICAL RESEARCH Mitral Regurgitation Augments Post-Myocardial Infarction Remodeling Failure of Hypertrophic Compensation Ronen Beeri, MD,* Chaim Yosefy, MD,* J. Luis Guerrero, BS,* Francesca Nesta, MD,* Suzan Abedat, MSC,§ Miguel Chaput, MD,* Federica del Monte, MD, PHD,† Mark D. Handschumacher, BS,* Robert Stroud, MS, Suzanne Sullivan, BS,* Thea Pugatsch, PHD,§ Dan Gilon, MD, FACC,§ Gus J. Vlahakes, MD, FACC,‡ Francis G. Spinale, MD, FACC, Roger J. Hajjar, MD, FACC,† Robert A. Levine, MD, FACC* Boston, Massachusetts; Jerusalem, Israel; and Charleston, South Carolina Objectives We examined whether mitral regurgitation (MR) augments post-myocardial infarction (MI) remodeling. Background MR doubles mortality after MI, but its additive contribution to left ventricular (LV) remodeling is debated and has not been addressed in a controlled fashion. Methods Apical MIs were created in 12 sheep, and 6 had an LV-to-left atrial shunt implanted, consistently producing re- gurgitant fractions of 30%. The groups were compared at baseline, 1, and 3 months. Results Left ventricular end-systolic volume progressively increased by 190% with MR versus 90% without MR (p 0.02). Pre-load–recruitable stroke work declined by 82 13% versus 25 16% (p 0.01) with MR, with de- creased remote-zone sarcoplasmic reticulum Ca 2 -ATPase levels (0.56 0.03 vs. 0.76 0.02, p 0.001), and decreased isolated myocyte contractility. In remote zones, pro-hypertrophic Akt and gp130 were upregulated in both groups at 1 month, but significantly lower and below baseline in the MR group at 3 months. Pro-apoptotic caspase 3 remained high in both groups. Matrix metalloproteinase (MMP)-13 and membrane-type MMP-1 were increased in remote zones of MR versus infarct-only animals at 1 month, then fell below baseline. The MMP tissue inhibitors rose from baseline to 3 months in all animals, rising higher in the MI MR–group border zone. Conclusions In this controlled model, moderate MR worsens post-MI remodeling, with reduced contractility. Pro-hypertrophic pathways are initially upregulated but subsequently fall below infarct-only levels and baseline; with sustained caspase 3 elevation, transformation to a failure phenotype occurs. Extracellular matrix turnover increases in MR animals. Therefore, MR can precipitate an earlier onset of dilated heart failure. (J Am Coll Cardiol 2008;51: 476–86) © 2008 by the American College of Cardiology Foundation Expansion of infarcted tissue begins acutely after myocardial infarction (MI). A more gradual remodeling process, how- ever, also involves the noninfarcted areas (1,2); initially compensatory, this process subsequently becomes mal- adaptive, generating a larger, more spherical, and poorly contracting ventricle (3–7), associated with reduced sur- vival (2,8,9). See page 487 Myocardial infarction can also cause mitral regurgitation (MR) by altering ventricular geometry and function (10– 12). Such ischemic MR doubles the risk of death after MI (13,14). Severe nonischemic MR has been shown to pro- mote left ventricular (LV) remodeling and reduce survival (15–19). However, whether more typically moderate isch- emic MR does so remains controversial. A recent paper maintained that “ischemic MR is a consequence, not a cause, of post-infarction remodeling,” because remodeling persisted despite annuloplasty with apparent MR reduction From the *Cardiac Ultrasound Laboratory, †Cardiovascular Research Center, and ‡Cardiac Surgery Department, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts; §Heart Institute and Cardiovascular Research Lab- oratory, Hadassah-Hebrew University Medical Center, Jerusalem, Israel; and the Cardiothoracic Surgery Department, Medical University of South Carolina, Charleston, South Carolina. Supported, in part, by a Grant-in-Aid (0350422N) of the American Heart Association; grants R01HL38176, R01HL72265, and K24HL67434 from the NIH/NHLBI; and a grant from the USA-Israel Binational Science Foundation (no. 2001037). Manuscript received May 3, 2007; revised manuscript received July 9, 2007, accepted July 16, 2007. Journal of the American College of Cardiology Vol. 51, No. 4, 2008 © 2008 by the American College of Cardiology Foundation ISSN 0735-1097/08/$34.00 Published by Elsevier Inc. doi:10.1016/j.jacc.2007.07.093

Transcript of Mitral Regurgitation Augments Post-Myocardial …Methods Apical MIs were created in 12 sheep, and 6...

Eieca

F‡So�CoKS

a

Journal of the American College of Cardiology Vol. 51, No. 4, 2008© 2008 by the American College of Cardiology Foundation ISSN 0735-1097/08/$34.00P

PRECLINICAL RESEARCH

Mitral Regurgitation AugmentsPost-Myocardial Infarction RemodelingFailure of Hypertrophic Compensation

Ronen Beeri, MD,* Chaim Yosefy, MD,* J. Luis Guerrero, BS,* Francesca Nesta, MD,*Suzan Abedat, MSC,§ Miguel Chaput, MD,* Federica del Monte, MD, PHD,†Mark D. Handschumacher, BS,* Robert Stroud, MS,� Suzanne Sullivan, BS,* Thea Pugatsch, PHD,§Dan Gilon, MD, FACC,§ Gus J. Vlahakes, MD, FACC,‡ Francis G. Spinale, MD, FACC,�Roger J. Hajjar, MD, FACC,† Robert A. Levine, MD, FACC*

Boston, Massachusetts; Jerusalem, Israel; and Charleston, South Carolina

Objectives We examined whether mitral regurgitation (MR) augments post-myocardial infarction (MI) remodeling.

Background MR doubles mortality after MI, but its additive contribution to left ventricular (LV) remodeling is debated and hasnot been addressed in a controlled fashion.

Methods Apical MIs were created in 12 sheep, and 6 had an LV-to-left atrial shunt implanted, consistently producing re-gurgitant fractions of �30%. The groups were compared at baseline, 1, and 3 months.

Results Left ventricular end-systolic volume progressively increased by 190% with MR versus 90% without MR (p �

0.02). Pre-load–recruitable stroke work declined by 82 � 13% versus 25 � 16% (p � 0.01) with MR, with de-creased remote-zone sarcoplasmic reticulum Ca2�-ATPase levels (0.56 � 0.03 vs. 0.76 � 0.02, p � 0.001), anddecreased isolated myocyte contractility. In remote zones, pro-hypertrophic Akt and gp130 were upregulated inboth groups at 1 month, but significantly lower and below baseline in the MR group at 3 months. Pro-apoptoticcaspase 3 remained high in both groups. Matrix metalloproteinase (MMP)-13 and membrane-type MMP-1 wereincreased in remote zones of MR versus infarct-only animals at 1 month, then fell below baseline. The MMPtissue inhibitors rose from baseline to 3 months in all animals, rising higher in the MI � MR–group borderzone.

Conclusions In this controlled model, moderate MR worsens post-MI remodeling, with reduced contractility. Pro-hypertrophicpathways are initially upregulated but subsequently fall below infarct-only levels and baseline; with sustainedcaspase 3 elevation, transformation to a failure phenotype occurs. Extracellular matrix turnover increases in MRanimals. Therefore, MR can precipitate an earlier onset of dilated heart failure. (J Am Coll Cardiol 2008;51:476–86) © 2008 by the American College of Cardiology Foundation

ublished by Elsevier Inc. doi:10.1016/j.jacc.2007.07.093

cv

(1(m(emc

xpansion of infarcted tissue begins acutely after myocardialnfarction (MI). A more gradual remodeling process, how-ver, also involves the noninfarcted areas (1,2); initiallyompensatory, this process subsequently becomes mal-daptive, generating a larger, more spherical, and poorly

rom the *Cardiac Ultrasound Laboratory, †Cardiovascular Research Center, andCardiac Surgery Department, Massachusetts General Hospital and Harvard Medicalchool, Boston, Massachusetts; §Heart Institute and Cardiovascular Research Lab-ratory, Hadassah-Hebrew University Medical Center, Jerusalem, Israel; and theCardiothoracic Surgery Department, Medical University of South Carolina,harleston, South Carolina. Supported, in part, by a Grant-in-Aid (0350422N)

f the American Heart Association; grants R01HL38176, R01HL72265, and24HL67434 from the NIH/NHLBI; and a grant from the USA-Israel Binationalcience Foundation (no. 2001037).

pManuscript received May 3, 2007; revised manuscript received July 9, 2007,

ccepted July 16, 2007.

ontracting ventricle (3–7), associated with reduced sur-ival (2,8,9).

See page 487

Myocardial infarction can also cause mitral regurgitationMR) by altering ventricular geometry and function (10–2). Such ischemic MR doubles the risk of death after MI13,14). Severe nonischemic MR has been shown to pro-ote left ventricular (LV) remodeling and reduce survival

15–19). However, whether more typically moderate isch-mic MR does so remains controversial. A recent paperaintained that “ischemic MR is a consequence, not a

ause, of post-infarction remodeling,” because remodeling

ersisted despite annuloplasty with apparent MR reduction

(tti

ttriirehtiuaf

cg(metiafittpamh

M

MtRmGiar(Cosbas2coicsoshA3oswpcnetacp3aascsoiws6ad(it

477JACC Vol. 51, No. 4, 2008 Beeri et al.January 29, 2008:476–86 MR Augments Post-MI Remodeling

20). Practically, “MR might not represent a target forherapy if indeed it is not the major culprit” (21), adding tohe uncertainty regarding the risk versus benefit of repairingschemic MR (22).

In patients, ischemic myocardial changes and regurgita-ion develop concurrently, precluding the ability to separateheir effects and analyze their interaction directly (23). Thisequires a model in which MR can be varied independentlyn the presence of MI, but without interventions such asnfarct patching (24–27) that might themselves influenceemodeling. Most experimental studies of post-MI remod-ling have used an infero-posterior MI model (27,28); thisas the disadvantage of linking the MI-induced remodelingo the development of MR. We therefore tested thencremental contribution of MR in a controlled mannersing a model in which the MI by itself does not cause MR,nd the MR can be created in a standardized and fixedashion (29,30).

In such a study, it must be recalled that remodeling is aomplex process, involving changes not only in ventriculareometry (3) and in global and regional myocardial function31), but also in calcium handling, molecular signals pro-oting hypertrophy, cardiomyocyte viability (32–34), and

xtracellular matrix turnover (35). Intracellular pathwayshat promote compensatory hypertrophy also tend, if chron-cally activated, to drive cells down pathways to cell death bypoptosis and thus to loss of contractile elements, withurther deterioration of cardiac function (Fig. 1). Accord-ngly, it is important to correlate evidence of remodeling athe molecular, cellular, and whole-heart levels (36). Weherefore tested the hypothesis that MR accompanying MIroduces greater remodeling than a comparable infarctlone, and may be associated with a different progression ofolecular events, with transition from signals promoting

ypertrophy to those inducing failure.

Figure 1 The Vicious Cycle of Remodeling

EC � extracellular; LV � left ventricle; MR � mitral regurgitation.

o

ethods

R model. A modification ofhe chronic MR sheep model ofankin et al. (29) was imple-ented in conjunction with Luisuerrero, a surgeon experienced

n physiological modeling, usingn 8-cm long, 8-mm diametereinforced Teflon (PTFE) graftEdwards Lifesciences, Irvine,alifornia) (cross-sectional areaf 0.50 cm2) implanted underterile conditions into the LVase and left atrial (LA) append-ge with intramuscular portionstiffened with epoxy resin (Fig.). The regurgitant flow wasonfirmed during each thoracot-my (see the following text) us-ng a transonic flow probe andolor Doppler echocardiography imaging. The standardizedhunt diameter and length have produced consistent levelsf MR (regurgitant fractions of approximately 30%, corre-ponding to moderate MR [37]). Animals were treated witheparin (3 days) and then oral aspirin.nimal studies. A total of 12 Dorsett hybrid sheep (20 to0 kg) were loaded for 3 days with amiodarone (200 mgrally twice a day) then anesthetized with thiopentothalodium (0.5 ml/kg), intubated, and ventilated at 15 ml/kgith a 2% isoflurane-oxygen mixture. All received glyco-yrrolate (0.4 mg intravenously) and prophylactic vancomy-in (0.5 g intravenously) and amiodarone (150 mg intrave-ous drip over the course of the operation). Surfacelectrocardiogram (ECG) was monitored, and a sterile lefthoracotomy performed with pericardial incision and cre-tion of a cradle. A high-fidelity micromanometer-tippedatheter (Millar Instruments Inc., Houston, Texas) waslaced into the LV. After baseline 2-dimensional (2D) and-dimensional (3D) echocardiography imaging, an antero-pical MI was produced by ligating the mid- to distal leftnterior descending coronary artery, known to produce aubstantial MI without MR (38). An immediate 2D echo-ardiography apical image was performed to confirm thateptal wall motion abnormality extended approximatelyne-third of the way from LV apex to base for standard-zation. Mitral regurgitation equivalent volume overloadas created for 90 days in 6 sheep with an open LV-to-LA

hunt (referred to as the infarct � MR group); in the other, the shunt was closed (infarct-only group). In addition tonalgesia, propranolol, 1 mg intravenously in 2 dividedoses, was given for evident stress and sinus tachycardia�150) upon extubation. Antibiotics (cephapirin, 0.5 gntravenously) and analgesics (buprenorphine, 0.3 mgwice a day) were administered for the next 5 days, and

Abbreviationsand Acronyms

EF � ejection fraction

LA � left atrium/atrial

LV � leftventricle/ventricular

MI � myocardial infarction

MMP � matrixmetalloproteinase

MR � mitral regurgitation

MT-MMP � membrane-typematrix metalloproteinase

SERCA2a � sarcoplasmicreticulum Ca2�-ATPase

TIMP � tissue inhibitor ofmatrix metalloproteinase

2D � 2-dimensional

3D � 3-dimensional

ral amiodarone (200 mg twice a

day) for the next 3.

cfcTArhtfAtDc3dcadnsrwtaqctfdppiMmvvraa

CscMtLmgewB(m(TDbcmwcoewbfl3cmMshcasCArbmfcpcam6P(PvapaP

478 Beeri et al. JACC Vol. 51, No. 4, 2008MR Augments Post-MI Remodeling January 29, 2008:476–86

During repeat sterile thoracotomy at day 30, 3D echo-ardiography was performed to evaluate LV remodeling andunction, with directed biopsies of the noninfarcted myo-ardium near and remote from the border zone using aruCut needle (Cook, Winston-Salem, North Carolina).t day 90, 3D echocardiography and blood sampling were

epeated at thoracotomy, followed by euthanasia and whole-eart pathological and myocyte size and contractility studieshat require considerable tissue. The animal studies con-ormed to National Institutes of Health Guidelines fornimal Research (Guide for the Care and Use of Labora-

ory Animals, National Research Council, Washington,C, 1996) and were approved by the institutional animal

are committee.D echocardiography and LV function. Three-imensional quantitation is necessary because LV shapehanges with chronic MR, as well as MI (39). Rotatedpical images were obtained at 10° intervals with an epicar-ial 5-MHz transesophageal echocardiography probe (So-os 7500, Philips Medical Systems, Andover, Massachu-etts), rotated by software and gated to ECG andespiration. Digital images were analyzed on a workstationith custom programs (40). Endocardial surfaces were

raced to calculate LV volumes and ejection fraction (EF),nd coded for wall motion abnormality to produce auantitative 3D wall motion map validated against a 36-rystal sonomicrometer array (41). Remodeling was quan-ified in terms of increasing LV volumes (42). Regurgitantraction was calculated as transonic MR-equivalent flowivided by LV ejection volume, with cross-correlation byulsed Doppler time–velocity integral of shunt flow multi-lied by shunt cross-sectional area. At thoracotomy, thenitial and final pressure–volume loops were obtained using

illar catheters and 4 sterile subendocardial crystals (Sono-etrics Corp., London, Ontario, Canada), with an inferior

ena cava occlusion to obtain the end-systolic pressure–olume relation for contractility (43) and pre-load–ecruitable stroke work (44). Crystals were located at the LVpex and base (between aorta and mitral annulus) and

Figure 2 Schematic and 2D EchocardiographicImage of the LA-LV Shunt Creating MR

Single arrows indicate flow direction; paired arrows indicate shunt exit fromthe LV. LA � left atrial; MI � myocardial infarction; 2D � 2-dimensional; otherabbreviations as in Figure 1.

nteriorly and posteriorly along a midventricular short-axis. s

rystals were not placed at day 30 to maximize sterility andurvival. dP/dt was obtained by the high-fidelity Millaratheter (Millar Instruments, Houston, Texas).

yocyte size and contraction. Small (5 to 10 g) areas ofissue were removed at euthanasia from the noninfarctedV and perfused via the corresponding coronary artery for 5in with low-magnesium Krebs-Henseleit (KH) cardiople-

ia solution. Myocardial cells were isolated using a well-stablished method (45,46). The LV sections were perfusedith Ca2�-free KH containing collagenase (Worthingtoniochem Corp., Lakewood, New Jersey) and hyaluronidase

Sigma, St. Louis, Missouri). Ventricular muscle tissue wasinced into small pieces and placed in enzyme buffer I

Sigma) in the presence of trypsin and deoxy-ribonuclease.he tissue was washed in a 3:1 mixture of Ca2�-free KH inulbecco’s phosphate-buffered saline (GIBCO, Gaithers-

urg, Maryland). Cells were loaded with the Ca2� fluores-ent indicator Fura-2 for 15 min in a light-sealed chamberounted on an inverted microscope. Myocyte length andidth were measured offline on 50 consecutive cells using

ommercial image analysis software to establish proportionsf normal short cells versus elongated cells characteristic ofccentric hypertrophy in the failing phenotype. The cellsere excited to contract using platinum electrodes in theath, and a dual excitation spectrofluorometer recordeduorescence emissions (505 nm from exciting wavelengths40 mm and 380 mm) to calculate intracellular calciumoncentrations. Cell motion was measured by a video-edgeotion detector (Crescent Electronics, Sandy, Utah) (45).olecular assays. We measured levels of several molecular

pecies that determine cell contractility, that modulate cellypertrophy and death, and that are responsible for extra-ellular matrix turnover. All protein assays were performednd specimens pooled from the same treatment group andtage.alcium cycling proteins: sarcoplasmic reticulum Ca2�-TPase (SERCA2a) and phospholamban. Sarcoplasmic

eticulum membranes were obtained from the myocardialiopsy specimens using a sucrose gradient centrifugationethod (47). Myocardium was homogenized and centri-

uged at 7,700 g for 20 min, and the supernatant filtered andentrifuged at 136,000 g for 60 min. The pellets, resus-ended and rehomogenized in 10% sucrose buffer, wereentrifuged at 55,000 g in a 10% to 40% sucrose gradient,nd the bands between 30% and 40% enriched in sarcoplas-ic reticulum were collected, centrifuged at 136,000 g for

0 min, and the pellet resuspended in freezing buffer.rotein was determined by a modified Bradford procedure

using bovine serum albumin for the standard curve).roteins were separated from the sarcoplasmic reticulumesicle preparation from tissue frozen in liquid nitrogen, andn immunoblot using monoclonal anti-SERCA2 and phos-holamban performed. Proteins levels were normalizedgainst total myocardial protein.ro-hypertrophic and anti-apoptotic cascades. We mea-

ured levels of Akt (protein kinase B) and gp130, which are

bicsoAapctaCMamM(pppmenat(TSdcNsanIm

R

RvateM0Rwibu�0ca(

m4CSrbaMcrIe�itbfEcrCgwEdalramba

479JACC Vol. 51, No. 4, 2008 Beeri et al.January 29, 2008:476–86 MR Augments Post-MI Remodeling

oth at their respective levels (cytosol and membrane)mportant crossroads in pro-hypertrophic signaling; andaspase 3, the final common pathway for intracellularignaling of apoptosis. Western blot analysis was performedn cell lysates from biopsies at baseline and days 30 and 90.ntibodies binding for anti-gp130, anti-phospho Akt, and

ntiactivated caspase 3 (48) were detected with aeroxidase-conjugated antimouse IgG and chemilumines-ence. �-Actin was used as a housekeeping control. Densi-ometry of the blots was assessed using commercially avail-ble software (Photoshop 5, Adobe Systems Inc., San Jose,alifornia).odulators of extracellular matrix turnover: matrix met-

lloproteinases (MMPs). Assays were performed forembrane-type matrix metalloproteinase-1 (MT1-MMP),MP-13, tissue inhibitor of matrix metalloproteinase

TIMP)-1, and TIMP-4 by immunoblotting using methodsreviously described (35). Left ventricular myocardial sam-les were homogenized and concentrated to prevent MMProteolytic activation and degradation. Trypsin (2.5 �M; 5in) was used to cleave nascent MMPs to fully activate

nzyme moieties and/or facilitate the removal of endoge-ous inhibitors. Relative TIMP-1, TIMP-4, MT1-MMP,nd MMP-13 levels were then measured by immunoblot-ing. A positive control was included in all immunoblotsMMP-13: CC068; MT1-MMP: CC1042) (Chemicon,emecula, California).tatistics. Statistical analysis was performed using a Stu-ent t test for continuous variables and a chi-square test forategorical variables (Stat View 5, SAS Institute Inc., Cary,orth Carolina). A value of p � 0.05 was considered

ignificant. Multiple comparisons were performed usingnalysis of variance (for repeated measurement whereeeded) and a Student-Neumann Keuls post-hoc test.nterobserver and intraobserver variability for 3D echo-easured LV volumes and EF in our laboratory is 3.5%.

esults

emodeling: LV volumes. Left ventricular end-diastolicolume relative to the acute post-MI value increased by anverage of 26% in the infarct-only group versus 130% inhose with volume overload (p � 0.02). Left ventricularnd-systolic volume progressively increased by 190% with

R-type volume overload versus 90% with MI only (p �.02) (Figs. 3 and 4).emodeling: LV function. Pre-load–recruitable strokeork from pressure-volume loops declined more in the

nfarct � MR group by 82 � 13% versus 25 � 16% ofaseline in the infarct-only group (p � 0.01). Left ventric-lar dP/dt also declined progressively in the MR group to 52

9% of baseline, but only by 22 � 12% in the shams (p �.04) (Fig. 4). These results were corroborated by reducedontractility of isolated cells with reduced calcium transientsnd increased diastolic intracellular calcium concentration

Tables 1 and 2). The diastolic relaxation constant tau was s

ore prolonged in the infarct � MR group (78 � 23 ms vs.4 � 13 ms, p � 0.01).alcium cycling proteins. As shown in Figure 5,ERCA2a abundance was downregulated in the infarctegion and border zone compared with the remote region inoth infarct � MR and infarct-only sheep LVs. There waslower level of SERCA2a in the remote zone of infarct �R than of infarct-only sheep. Phospholamban was un-

hanged in all regions. These changes are typical foremodeling myocardium.ntracellular signaling pathways. Akt, a pro-hypertrophicnzyme (49), was upregulated in the remote zones of infarct

MR and infarct-only animals at 1 month, but was lowern animals with than without MR at 3 months (Fig. 6). Inhe infarct � MR group at 3 months, Akt also fell belowaseline of the same group. The same pattern was observedor the interleukin-6-family receptor subunit gp130 (50).xpression of activated caspase 3 (32–34,51), the final

ommon pathway of apoptosis, remained elevated in allegions of both groups at both 1 and 3 months.

ell morphology. Myocytes in the MR group were elon-ated, characteristic of myocardial failure (45), comparedith cells from the MI-only animals (Fig. 7).xtracellular matrix and MMPs. In the remote myocar-ium of infarct � MR sheep, at initial follow-up, there wassignificant increase in MMP-13 and MT1-MMP, fol-

owed by a decrease at the time of sacrifice (Fig. 8, “shuntemote” bars): MMP-13 increased to 238 � 29% of baselinet follow-up, and then fell to 79 � 13% of baseline at 3onths sacrifice; MT1-MMP increased to 165 � 23% of

aseline at follow-up, and then fell to 85 � 16% of baselinet sacrifice. In the remote myocardium of infarct-only

Figure 3 Echocardiographic 3D Reconstructionsof the Endocardial Surface of the LV

Echocardiographic 3-dimensional (3D) reconstructions of the endocardial sur-face of infarct (MI) only (A and B) and infarct � MR (C and D) animals at dias-tole (top) and systole (bottom). Note the enlarged and spherical infarct � MRventricles. Abbreviations as in Figures 1 and 2.

heep, the rise and fall of MMP-13 did not reach statistical

s�i

bcbgbbhzTsg

D

MloabtaatTepoicus(mriI

IC

*r

IM

*

480 Beeri et al. JACC Vol. 51, No. 4, 2008MR Augments Post-MI Remodeling January 29, 2008:476–86

ignificance (170 � 85% of baseline at initial follow-up; 7522% at sacrifice), and MT1-MMP did not significantly

Figure 4 Measurement of LV Volumes and Function

End-diastolic volume (EDV) (A) and end-systolic volume (ESV) (B) of infarct-only(yellow bars) and infarct � MR (red bars) sheep at baseline (pre), 1 monthfollow-up (f/u), and 3 months sacrifice. (C and D) dP/dt and pre-load–recruit-able stroke work (PRSW) of infarct-only (broken lines) and infarct � MR (solidlines) sheep at pre, f/u, and 3 months sacrifice. *p � 0.05. Abbreviations asin Figure 1.

ncrease at 1 month (Fig. 8, “sham remote” bars). In ther

order zones of the infarct-only sheep, there was a signifi-ant decrease in MT1-MMP at sacrifice (to 60 � 8% ofaseline), with levels maintained in the infarct � MRroup. In contrast, TIMP-4 showed progressive rises fromaseline to initial follow-up and then to sacrifice in theorder and remote zones of both groups, with significantlyigher levels at sacrifice in the infarct � MR group borderone (207 � 9% vs. 152 � 10% of baseline, p � 0.05).issue inhibitor of matrix metalloproteinase-1 showed a

imilar pattern of progressive increases in the infarct � MRroup remote zone, with a significant late rise at sacrifice.

iscussion

yocardial infarction, with loss of contracting myocytes,eads to a sequence of events aimed at preserving cardiacutput, including increased LV end-diastolic volume tougment pre-load. Cells in noninfarcted areas hypertrophyy addition of contractile elements in series, creating eccen-ric hypertrophy without increased wall thickness. Thisllows the LV to dilate, a process that is also promoted byltered MMP activation with extracellular matrix degrada-ion (52), and acceleration of cell death by apoptosis (32,53).his process eventually culminates in systolic enlargement,

xtensive fibrosis, and LV failure, the extent of whichredicts further cardiac events (2,9). Volume and pressureverload increase wall stress, which can aggravate remodel-ng (54) and promote eccentric hypertrophy through in-reased fiber stretch (Fig. 1). Conversely, hemodynamicallynloading the LV reverses the functional and cellulartigmata of cardiac failure as well as the remodeling process55,56). Neurohumoral activation and activation of inflam-atory mediators and cytokines initially compensate for

educed cardiac output but, in the long term, mediatencreased load, cell death, and cardiac fibrosis (50,57–59).ncreased wall stress also up-regulates the production and

solated Cardiomyocyteontractility in the Different Groups

Table 1 Isolated CardiomyocyteContractility in the Different Groups

ContractilityMeasurements

NormalControl Sheep Infarct � MR Infarct Only

Cell shortening (%) 5.7 � 0.4 2.6 � 0.7* 3.5 � 0.3*†

t90% (ms) 390 � 82 676 � 68* 510 � 59*†

p � 0.01 compared with normal control sheep; †p � 0.01 compared with infarct � mitralegurgitation (MR) group.

t90% � time to 90% relaxation.

ntracellular Calciumeasurements in the Different Groups

Table 2 Intracellular CalciumMeasurements in the Different Groups

CalciumMeasurements

NormalControl Sheep Infarct � MR Infarct Only

Diastolic (Ca2�) (nM) 230 � 41 320 � 21* 275 � 22*†

Systolic (Ca2�) (nM) 796 � 24 535 � 44* 714 � 43*‡

t90% (ms) 361 � 34 542 � 38* 455 � 43*†

p � 0.01 compared with normal control sheep; †p � 0.03 compared with infarct � mitral

egurgitation (MR) group; ‡p � 0.005 compared with infarct � MR group.

t90% � time to 90% relaxation of the calcium signal.

ravda

gto(tefAlecdcbpc(

prbvp

iiMdniaac

paraspfd

mcMptacmw

481JACC Vol. 51, No. 4, 2008 Beeri et al.January 29, 2008:476–86 MR Augments Post-MI Remodeling

elease of MMPs, which are believed to be pivotal inltering the extracellular matrix in ways that promoteentricular dilatation (60). Their transgenic activation in-uces congestive heart failure (61), and their inhibitionttenuates post-MI remodeling (62,63).

Mitral regurgitation, caused by alterations in ventriculareometry and function after MI (10,12), can itself initiatehe remodeling cascade, and causes progressive deteriorationf ventricular function at a cellular and molecular level15–17). Mitral regurgitation considerably alters the load onhe LV. It increases diastolic wall stress, which can induceccentric LV hypertrophy and subsequent dilatation andailure (18), and it thereby increases early systolic wall stress.lthough MR permits LV emptying during systole into the

ower-pressure LA, it has been shown actually to increasend-systolic wall stress and, hence, afterload in patients withhronic MR (64,65). Moreover, MR induces further LVilatation due to activation of neurohumoral and cytokineomponents of the remodeling cascade (66–68). As MR isoth a cause and a result of LV remodeling, it canotentially exacerbate the vicious cycle spiraling down toardiac failure unless the remodeling or the MR is reversed69–71).

Regarding clinical implications, knowing whether thearallel occurrence of MR and MI causes more pronouncedemodeling is critical, because at least MR can be eliminatedy repairing or replacing the valve, and thus relieving theolume overload induced by it. Separating these 2 dynamic

Figure 5 SERCA2a and Phospholamban Quantification

(A) Quantification of sarcoplasmic reticulum Ca2�-ATPase (SERCA2a) activity in reImmunoblots from different regions of the myocardium in different groups for SERCdium that was homogenized and protein extracted. Abbreviations as in Figures 1 a

rocesses in an experimental model is a challenge, because o

n most existing models they are linked. One recent study ofnferior MIs, in fact, concluded that prevention of ischemic

R does not influence the outcome of remodeling (20),espite benefits in nonischemic models (18) and concerns inonischemic patients (72). In our model, we therefore

mplanted an LV-to-LA shunt, which created a moderatend standardized MR-like regurgitant flow, independent ofmedium-sized antero-apical MI that by itself does not

ause MR.In this controlled model, moderate MR worsened

ost-MI remodeling. Specifically, it induced large diastolicnd systolic LV volumes reflecting volume overload andeduced global function. Reduced contractility was alsopparent in less pre-load–dependent physiological measuresuch as pre-load–recruitable stroke work. These changesaralleled decreased single-cell contractility in the nonin-arcted myocardium with reduced SERCA2a levels, whichirectly correlate with cell contraction (46).In contrast to progressive changes in the LV, severalediators of the hypertrophic process underwent biphasic

hanges in the noninfarcted myocardium of animals withR-type volume overload. These include gp130, a glyco-

rotein that forms heterodimers to produce different recep-ors, notably of the interleukin 6/CT1/LIF family. Thectivation of these receptors has been linked to cardiomyo-yte hypertrophy (50,73), and their reduced abundance,anifested by reduced gp130 levels, has been associatedith the transition from hypertrophy to failure (74,75). Akt,

border, and infarct regions of experimental and normal control sheep hearts. (B)nd phospholamban. Note that in the infarct region, there was residual myocar-

mote,A2a and 2.

r protein kinase B, is a serine-threonine kinase activated by

scrpocltpiarwhmrcdp

towmwfmoh

MzlHfiodilhdssaoncp

ais

482 Beeri et al. JACC Vol. 51, No. 4, 2008MR Augments Post-MI Remodeling January 29, 2008:476–86

everal pro-survival and -hypertrophic factors, among othersardiotrophin-1 (76), acting through the gp130-containingeceptor, or growth factor receptors activatinghosphoinositide-3 kinase (77).The constitutive activationf Akt in a model of rat ischemia-reperfusion injury reducedell death and improved function (78). Like gp130, reducedevels of Akt are related to increased apoptosis and aransition from compensatory hypertrophy to a failurehenotype (49). In our model, while both infarct � MR andnfarct-only sheep had elevated expression of Akt and gp130fter 1 month, the MR sheep demonstrated a notableeduction in these protein levels after 3 months, comparedith the no-MR animals and with baseline. The pro-ypertrophic process, which begins in both groups at 1onth, continues in the animals without MR but is down-

egulated at 3 months in the MR group, in association withontinued adverse remodeling and LV dysfunction. Thisecline may represent earlier termination of the hypertro-

Figure 6 Protein Levels of Akt, gp130, and Caspase 3

Protein expression of Akt, gp130, and activated caspase 3 from infarct-only(broken lines) and infarct � mitral regurgitation (solid lines) sheep at base-line, 1 month follow-up (f/u), and 3 months sacrifice. Note the biphasic changein infarct � mitral regurgitation animals for gp130 and Akt expression at fol-low-up (high) and sacrifice, while no significant change is noted in caspase 3levels. Values are � standard error of the mean, *p � 0.01.

hic process or transition to a failure phenotype specific to

he persistent-MR situation; either way, it represents failuref the hypertrophic process. The ongoing drive to cell death,ith the suppressed pro-hypertrophic drive, further pro-otes the transition to failure. These findings are consistentith the emerging view of LV remodeling in general as a

ailed attempt at compensation for initial stresses on theyocardium—in this case, exacerbated by the MR volume

verload (7,74); in others, it reflects the transition fromypertensive hypertrophy to failure (79,80).There was also excessive activation of MMPs, notablyMP-13 and MT-MMP1, in the noninfarcted remote

one of the MR sheep. This could induce excess extracel-ular matrix turnover and potentially exacerbate remodeling.

owever, these same MMP levels fell at sacrifice, with theall in remote-zone MT-MMP1 being significantly greatern the MR sheep, and the fall in MMP-13 being significantnly for the MR group. As this change parallels theown-regulation of the pro-hypertrophic signaling process,t could be also part of the failure of compensation byimiting the adaptive geometrical changes in myocardialypertrophy. Matrix metalloproteinase inhibitors showed aifferent pattern, with a progressive rise from baseline toacrifice that was significantly greater with MR. Thisuggests a possible shift from matrix turnover to matrixccumulation and stabilization over time, a response typicalf remodeling myocardium (81). However, it is clear thateither the MMP nor the TIMP changes adequatelyompensate for the remodeling process, as LV volumerogressively rose while systolic function fell.This study has several limitations. Ischemic MR affectingnative valve often progressively increases (28,69–71), but

s inherently linked to the underlying MI and is nottandardized. Based on the motivation for this study, it was

Figure 7 Photomicrographs of Separated Cardiomyocytes

Elongated myocytes from sheep with MI and MR shunt (A) versus more normalcells from MI-only animals (B). Note the different scales used to accommodatethe elongated cells of the MI � MR sheep within the field. Abbreviations as inFigures 1 and 2.

cgdptot

ciMra

iprt

trhprga

483JACC Vol. 51, No. 4, 2008 Beeri et al.January 29, 2008:476–86 MR Augments Post-MI Remodeling

ritical to separate the 2 processes of infarction and regur-itation to determine the incremental role of MR mostirectly, and to do so with a standardized orifice, whichrovided stable regurgitant fractions of �30% throughouthe study. Although the LV also fills through this valvelessrifice, based on prior experience, this should not impor-antly alter LV filling (29).

We did not assess whether any one of the molecularhanges reported in the present study is causative. Wentended to test whether differences in remodeling based on

R are associated with molecular changes described in theemodeling process. Determining which factors are caus-

Figure 8 MMP Abundance

Levels of membrane-type (MT) matrix metalloproteinase (MMP)1 and tissue inhibitinfarct � mitral regurgitation (red bars) animals at first follow-up (T2) and sacrifice� mitral regurgitation relative to myocardial infarction only animals for the same z

tive versus concomitants, and studying all possible factors, c

s beyond the scope of this study. Nevertheless, we analyzedathways extensively linked to the pro-hypertrophic andemodeling processes, and causally implicated throughransgenic and pharmacologic modification (61,74,78).

While the impact of isolated MR is important to explore,he study specifically addresses the challenge posed by theecent literature (20), which can be stated as an alternativeypothesis that remodeling in ischemic MR is caused only orredominantly by the underlying infarct, not the MR. Thisequired comparing infarct-only versus infarct � MRroups. The data do show important effects of the infarctlone, with end-systolic volume nearly doubling, and in-

atrix metalloproteinase (TIMP)-4 in the infarct-only (“sham”, green bars) andime points. *p � 0.05 relative to baseline; #p � 0.05 for myocardial infarctiond stage; $p � 0.05 for sacrifice relative to initial follow-up.

or of m(T3) t

one an

reases in gp130, Akt, and caspase. The changes with infarct

pnbmapire

biaTsctbesiTtrrtpfrocMMcTofro

C

Tdipmufpfpwifm

ttt

RCHr

R

1

1

1

1

1

1

1

1

484 Beeri et al. JACC Vol. 51, No. 4, 2008MR Augments Post-MI Remodeling January 29, 2008:476–86

lus MR, however, were greater, with end-systolic volumeearly tripling, and a different pattern of activation followedy exhaustion of hypertrophic- and apoptotic-signalingolecules gp130 and Akt. The findings, therefore, suggest

n additive rather than exclusive role of MR. This is sup-orted by our data (unpublished results) from sheep with andentical shunt and no MI, which did not cause significantemodeling in the same 3-month time-frame in terms ofither volume or contractility changes.

Recently, contribution of MR to post-MI remodeling haseen challenged by Guy et al. (20) in an inferior MI modeln which either MR was reduced using an undersizednnuloplasty ring repair, or MI was restrained using a mesh.he MR repair group remodeled more, leading to the

uggestion that the MR is not causative, but may be aonsequence of post-MI remodeling. The main caveat tohese conclusions is that undersized ring annuloplasty mayy itself alter LV mechanics, as recently shown (82), so as toxacerbate remodeling. Thus, the excess remodeling ob-erved may have been caused, at least partially, by the repairtself, which was performed 2 weeks before infarct creation.he difficulty in eliminating MR as a potential contributor

o continued remodeling by annuloplasty has also beeneported (72). Another recent paper (83) has suggested thatepairing MR in a subgroup of patients with LV dysfunc-ion has no survival benefit. While this is intriguing, theatient population in that study had prominent LV dys-unction at baseline (�30% EF), and thus it is possible thatepair was performed too late for many to benefit in termsf LV function or outcome. Based on their inclusionriteria, the patients also had at least moderate-to-severe

R—more severe than in our model. Further, unlike thisI model, roughly 40% of the patients did not have

oronary artery disease as the basis for their LV dysfunction.hus, these clinical data cannot readily be compared withur relatively mild MI model with normal baseline LVunction and moderate MR. There is definitely a need for aandomized controlled study to clinically address the issuef mitral valve repair in moderate ischemic MR.

onclusions

his study demonstrates that MR accompanying MI pro-uces greater ventricular remodeling than a comparablenfarction alone, with an earlier transition to a failurehenotype. This is the case even for only moderate MR,ost typical of the ischemic situation, for which the greatest

ncertainty exists regarding impact on the heart and needor intervention. The changes at the level of the whole heartarallel cellular and molecular abnormalities in the nonin-arcted myocardium that reflect the complex remodelingrocess. These molecular events also progress differentlyith MR than with comparable infarction alone, with an

nitial rise in pro-hypertrophic and antiapoptotic signals,ollowed by their exhaustion. Recognizing this pattern can

otivate a search for the responsible molecular “switch,” so

1

hat decompensation might be limited or reversed, avertinghe earlier onset of dilated heart failure precipitated by MRhat may portend a worse prognosis.

eprint requests and correspondence: Dr. Robert A. Levine,ardiac Ultrasound Laboratory YAW5, Massachusetts Generalospital, 55 Fruit Street, Boston, Massachusetts 02114. E-mail:

[email protected].

EFERENCES

1. Pfeffer MA, Braunwald E. Ventricular remodeling after myocardialinfarction: experimental observations and clinical implications. Circu-lation 1990;81:1161–72.

2. Cohn JN, Ferrari R, Sharpe N. Cardiac remodeling—concepts andclinical implications: a consensus paper from an international forum oncardiac remodeling. J Am Coll Cardiol 2000;35:569–82.

3. Picard MH, Wilkins GT, Ray PA, Weyman AE. Progressive changesin ventricular structure and function during the year after acutemyocardial infarction. Am Heart J 1992;124:24–31.

4. Pfeffer MA. Left ventricular remodeling after acute myocardial infarc-tion. Annu Rev Med 1995;46:455–66.

5. Gianuzzi P, Temporelli PL, Bosimi E, et al., for the Gruppo Italianoper lo Studio della Sopravvivenza nell’Infarto Miocardico-3 EchoSubstudy Investigators. Heterogeneity of left ventricular remodelingafter acute myocardial infarction: results of the Gruppo Italiano per loStudio della Sopravvivenza nell’Infarto Miocardico-3 echo substudy.Am Heart J 2001;141:131–8.

6. Davidoff R, Picard MH, Force T, et al. Spatial and temporal variabilityin the pattern of recovery of ventricular geometry and function afteracute occlusion and reperfusion. Am Heart J 1994;127:1231–41.

7. Mann DL, Bristow MR. Mechanisms and models in heart failure. Thebiomechanical model and beyond. Circulation 2005;111:2837–49.

8. Lamas GA, Mitchell GF, Flaker GC, et al. Clinical significance ofmitral regurgitation after acute myocardial infarction. Circulation1997;96:827–33.

9. White HD, Norris RM, Brown MA, Brandt PW, Whitlock RM,Wild CJ. Left ventricular end-systolic volume as the major determi-nant of survival after recovery from myocardial infarction. Circulation1987;76:44–51.

0. Otsuji Y, Handschumacher MD, Schwammethal E, et al. Insightsfrom three-dimensional echocardiography into the mechanisms offunctional mitral regurgitation: direct in vivo demonstration of alteredleaflet geometry. Circulation 1997;96:1999–2008.

1. Cochran RP, Kunzelman KS. Effect of papillary muscle position onmitral valve function: relationship to homografts. Ann Thorac Surg1998;66:S155–61.

2. Kaul S, Spotnitz WD, Glasheen WP, Touchstone DA. Mechanism ofischemic mitral regurgitation: an experimental evaluation. Circulation1991;84:2167–80.

3. Lehmann KG, Francis CK, Dodge HT, the TIMI Study Group.Mitral regurgitation in early myocardial infarction: incidence, clinicaldetection and prognostic implications. Ann Intern Med 1992;117:10–7.

4. Grigioni F, Enriquez-Sarano M, Zehr KJ, Bailey KR, Tajik AJ.Ischemic mitral regurgitation: long-term outcome and prognosticimplications with quantitative Doppler assessment. Circulation 2001;103:1759–64.

5. Carabello BA, Nakano K, Corin W, Biederman R, Spann JF Jr. Leftventricular function in experimental volume overload hypertrophy.Am J Physiol 1989;256:H974–81.

6. Urabe Y, Mann DL, Kent RL, et al. Cellular and ventricularcontractile dysfunction in experimental canine mitral regurgitation.Circ Res 1992;70:131–47.

7. Ishihara K, Zile MR, Kanazawa S, et al. Left ventricular mechanicsand myocyte function after correction of experimental chronic mitralregurgitation by combined mitral valve replacement and preservationof the native mitral valve apparatus. Circulation 1992;86:16–25.

8. Spinale FG, Ishihra K, Zile MR, DeFryte G, Crawford FA, CarabelloBA. Structural basis for changes in left ventricular function and

1

2

2

2

2

2

2

2

2

2

2

3

3

3

3

3

3

3

3

3

3

4

4

4

4

4

4

4

4

4

4

5

5

5

5

5

5

5

5

5

5

6

6

6

485JACC Vol. 51, No. 4, 2008 Beeri et al.January 29, 2008:476–86 MR Augments Post-MI Remodeling

geometry because of chronic mitral regurgitation and after correctionof volume overload. J Thorac Cardiovasc Surg 1993;106:1147–57.

9. Ling LH, Enriquez-Sarano M, Seward JB, et al. Clinical outcomeof mitral regurgitation due to flail leaflet. N Engl J Med 1996;335:1417–23.

0. Guy TS, Moainie SL, Gorman JH, et al. Prevention of ischemic mitralregurgitation does not influence the outcome of remodeling afterposterolateral myocardial infarction. J Am Coll Cardiol 2004;43:377–83.

1. Carabello BA. Ischemic mitral regurgitation and ventricular remodel-ing. J Am Coll Cardiol 2004;43:384–5.

2. Trichon BH, Glower DD, Shaw LK, et al. Survival after coronaryrevascularization, with and without mitral valve surgery, in patientswith ischemic mitral regurgitation. Circulation 2003;108 Suppl 1:103–10.

3. Neskovic AN, Marinkovic J, Bojic M, Popovic AD. Early mitralregurgitation after acute myocardial infarction does not contribute tosubsequent left ventricular remodeling. Clin Cardiol 1999;22:91–4.

4. Oh JH, Badhwar V, Mott BD, Li CM, Chiu RC. The effects ofprosthetic cardiac binding and adynamic cardiomyoplasty in a model ofdilated cardiomyopathy. J Thorac Cardiovasc Surg 1998;116:148–53.

5. Kelley ST, Malekan R, Gorman JH III, et al. Restraining infarctexpansion preserves left ventricular geometry and function after acuteanteroapical infarction. Circulation 1999;99:135–42.

6. Hung J, Guerrero JL, Handschumacher MD, Supple G, Sullivan S,Levine RA. Reverse ventricular remodeling reduces ischemic mitralregurgitation: echo-guided device application in the beating heart.Circulation 2002;106:2594–600.

7. Moainie SL, Guy TS, Gorman JH, et al. Infarct restraint attenuatesremodeling and reduces chronic ischemic mitral regurgitation afterpostero-lateral infarction. Ann Thorac Surg 2002;74:444–9.

8. Llaneras MR, Nance ML, Streicher JT, et al. Large animal model ofischemic mitral regurgitation. Ann Thorac Surg 1994;57:432–9.

9. Rankin JS, Nicholas LM, Kouchoukos NT. Experimental mitralregurgitation: effects on left ventricular function before and afterelimination of chronic regurgitation in the dog. J Thorac CardiovascSurg 1975;70:478–88.

0. Braunwald E, Welch GH Jr., Sarnoff SJ. Hemodynamic effects ofquantitatively varied experimental mitral regurgitation. Circ Res 1957;5:539–45.

1. Fisher JP, Picard MH, Mikan JS, et al. Quantitation of myocardialdysfunction in ischemic heart disease by echocardiographic endocardialsurface mapping: correlation with hemodynamic status. Am Heart J1995;129:1114–21.

2. Cheng W, Kajstura J, Nitahara JA, et al. Programmed myocyte celldeath affects the viable myocardium after infarction in rats. Exp CellRes 1996;226:316–27.

3. Foo RS, Mani K, Kitsis RN. Death begets failure in the heart. J ClinInvest 2005;115:565–71.

4. Wencker D, Chandra M, Nguyen K, et al. A mechanistic role forcardiac myocyte apoptosis in heart failure. J Clin Invest 2003;111:1497–504.

5. Spinale FG, Coker ML, Heung LJ, et al. A matrix metalloproteinaseinduction/activation system exists in the human left ventricular myo-cardium and is upregulated in heart failure. Circulation 2000;102:1944–9.

6. Sabbah HN, Sharov VG, Gupta RC, et al. Reversal of chronicmolecular and cellular abnormalities due to heart failure by passivemechanical ventricular containment. Circ Res 2003;93:1095–101.

7. Zoghbi WA, Enriquez-Sarano M, Foster E, et al. Recommendationsfor evaluation of the severity of native valvular regurgitation withtwo-dimensional and Doppler echocardiography. J Am Soc Echocar-diogr 2003;16:777–82.

8. Gorman JH, Gorman RC, Plappert T, et al. Infarct size and locationdetermine development of mitral regurgitation in the sheep model.J Thorac Cardiovasc Surg 1998;115:615–22.

9. Young AA, Orr R, Smaill BH, Dell’Italia LJ. Three-dimensionalchanges in left and right ventricular geometry in chronic mitralregurgitation. Am J Physiol 1996;271:H2689–700.

0. Handschumacher MD, Lethor J-P, Siu SC, et al. A new integratedsystem for three-dimensional echocardiographic reconstruction: devel-opment and validation for ventricular volume with application in

human subjects. J Am Coll Cardiol 1993;21:743–53.

1. Buck T, Handschumacher MD, Tanabe H, Guerrero JL, Levine RA.A new method for automatic quantification and visualization ofregional left ventricular dysfunction by three-dimensional echocardi-ography: direct in vivo validation against three-dimensional sonomi-crometry (abstr). J Am Coll Cardiol 1999;33:440A.

2. Mitchell GF, Lamas GA, Vaughan DE, Pfeffer MA. Left ventricularremodeling in the year after first anterior myocardial infarction: aquantitative analysis of contractile segment lengths and ventricularshape. J Am Coll Cardiol 1992;19:1136–44.

3. Nakano K, Swindle MM, Spinale FG, et al. Depressed contractilefunction due to canine mitral regurgitation improves after correction ofthe volume overload. J Clin Invest 1991;87:2153–61.

4. Glower DD, Spratt JA, Snow ND, et al. Linearity of the Frank-Starling relationship in the intact heart: the concept of preloadrecruitable stroke work. Circulation 1985;71:994–1009.

5. del Monte F, O’Gara P, Poole-Wilson PA, Yacoub M, Harding SE.Cell geometry and contractile abnormalities of myocytes from failinghuman left ventricle. Cardiovasc Res 1995;30:281–90.

6. del Monte F, Harding SE, Schmidt U, et al. Restoration of contractilefunction in isolated cardiomyocytes from failing human hearts by genetransfer of SERCA2a. Circulation 1999;100:2308–11.

7. Chu A, Dixon MC, Saito A, Seiler S, Fleischer S. Isolation ofsarcoplasmic reticulum fractions referable to longitudinal tubules andjunctional terminal cisternae from rabbit skeletal muscle. MethodsEnzymol 1988;157:36–46.

8. Molkentin JD, Dorn GW II. Cytoplasmic signaling pathways thatregulate cardiac hypertrophy. Annu Rev Physiol 2001;63:391–426.

9. Haq S, Choukroun G, Lim HW, et al. Differential activation of signaltransduction pathways in human hearts with hypertrophy versusadvanced heart failure. Circulation 2001;103:670–7.

0. Hirota H, Yoshida K, Kishimoto T, Taga T. Continuous activation ofgp130, a signal-transducing receptor component for interleukin6-related cytokines, causes myocardial hypertrophy in mice. Proc NatlAcad Sci U S A 1995;92:4862–6.

1. Communal C, Sumandea M, de Tombe P, Narula J, Solaro RJ, HajjarRJ. Functional consequences of caspase activation in cardiac myocytes.Proc Natl Acad Sci U S A 2002;99:6252–6.

2. Spinale FG, Coker ML, Thomas CV, Walker JD, Mukherjee R,Hebbar L. Time-dependent changes in matrix metalloproteinaseactivity and expression during the progression of congestive heartfailure: relation to ventricular and myocyte function. Circ Res 1998;82:482–95.

3. Sam F, Sawyer DB, Chang DLF, et al. Progressive left ventricularremodeling and apoptosis late after myocardial infarction in mouseheart. Am J Physiol 2000;279:H422–8.

4. Rumberger JA. Ventricular dilatation and remodeling after myocardialinfarction. Mayo Clin Proc 1994;69:664–74.

5. Levin HR, Oz MC, Chen JM, Packer M, Rose EA, Burkhoff D.Reversal of chronic ventricular dilation in patients with end-stagecardiomyopathy by prolonged mechanical unloading. Circulation1995;91:2717–20.

6. Heerdt PM, Holmes JW, Cai B, et al. Chronic unloading by leftventricular assist device reverses contractile dysfunction and alters geneexpression in end-stage heart failure. Circulation 2000;102:2713–9.

7. Ono K, Matsumori A, Shioi T, Furukawa Y, Sasayama S. Cytokinegene expression after myocardial infarction in rat hearts: possibleimplication in left ventricular remodeling. Circulation 1998;98:149–56.

8. Wollert KC, Taga T, Saito M, et al. Cardiotrophin-1 activates adistinct form of cardiac muscle cell hypertrophy. J Biol Chem1996;271:9535–45.

9. Sivasubramanian N, Coker ML, Kurrelmeyer KM, et al. Left ventric-ular remodeling in transgenic mice with cardiac restricted overexpres-sion of tumor necrosis factor. Circulation 2001;104:826–31.

0. MacKenna D, Summerour SR, Villarreal FJ. Role of mechanicalfactors in modulating cardiac fibroblast function and extracellularmatrix synthesis. Cardiovasc Res 2000;46:257–63.

1. Kim HE, Dalal SS, Young E, Legato MJ, Weisfeldt ML, D’ArmientoJ. Disruption of the myocardial extracellular matrix leads to cardiacdysfunction. J Clin Invest 2000;106:857–66.

2. Coker ML, Thomas CV, Clair MJ, et al. Myocardial matrix metallo-proteinase activity and abundance with congestive heart failure. Am J

Physiol 1998;274:H1516–23.

6

6

6

6

6

6

6

7

7

7

7

7

7

7

7

7

7

8

8

8

8

486 Beeri et al. JACC Vol. 51, No. 4, 2008MR Augments Post-MI Remodeling January 29, 2008:476–86

3. Peterson JT, Hallak H, Johnson L, et al. Matrix metalloproteinaseinhibition attenuates left ventricular remodeling and dysfunction in arat model of progressive heart failure. Circulation 2001;103:2303–9.

4. Corin WJ, Monrad ES, Murakami T, Nonogi H, Hess OM, Kray-enbuehl HP. The relationship of afterload to ejection performance inchronic mitral regurgitation. Circulation 1987;76:59–67.

5. Zile MR, Gaasch WH, Levine HT. Left ventricular stress-dimension-shortening relations before and after correction of chronic aortic andmitral regurgitation. Am J Cardiol 1985;56:99.

6. Dell’Italia LJ, Meng QC, Balcells E, et al. Increased ACE andchymase-like activity in cardiac tissue of dogs with chronic mitralregurgitation. Am J Physiol 1995;269:H2065–73.

7. Kapadia SR, Yakoob K, Nader S, Thomas JD, Mann DL, Griffin BP.Elevated circulating levels of serum tumor necrosis factor-alpha inpatients with hemodynamically significant pressure and volume over-load. J Am Coll Cardiol 2000;36:208–12.

8. Talwar S, Squire IB, Davies JE, Ng LL. The effect of valvularregurgitation on plasma cardiotrophin-1 in patients with normal leftventricular systolic function. Eur J Heart Fail 2000;2:387–91.

9. Otsuji Y, Handschumacher MD, Liel-Cohen N, et al. Mechanism ofischemic mitral regurgitation with segmental left ventricular dysfunction:three-dimensional echocardiographic studies in models of acute andchronic progressive regurgitation. J Am Coll Cardiol 2001;37:641–8.

0. Liel-Cohen N, Guerrero JL, Otsuji Y, et al. Design of a new surgicalapproach for ventricular remodeling to relieve ischemic mitral regur-gitation. Circulation 2000;101:2756–63.

1. Hung J, Papakostas L, Tahta SA, et al. Mechanism of recurrentischemic mitral regurgitation post-annuloplasty: continued LV remod-eling as a moving target. Circulation 2004;110:85–90.

2. Enriquez-Sarano M, Avierinos JF, Messika-Zeitoun D, et al. Quan-titative determinants of the outcome of asymptomatic mitral regurgi-tation. N Engl J Med 2005;352:875–83.

3. Kunisada K, Tone E, Fujio Y, Matsui H, Yamauchi-Takihara K,Kishimoto T. Activation of gp130 transduces hypertrophic signals via

STAT3 in cardiac myocytes. Circulation 1998;98:346–52.

4. Hirota H, Chen J, Betz UAK, et al. Loss of a gp130 cardiac muscle cellsurvival pathway is a critical event in the onset of heart failure duringbiomechanical stress. Cell 1999;97:189–98.

5. Zolk O, Ng LL, O’Brien RJ, Weyand M, Eschenhagen T. Aug-mented expression of cardiotrophin-1 in failing human hearts isaccompanied by diminished glycoprotein 130 receptor protein abun-dance. Circulation 2002;106:1430–2.

6. Craig R, Wagner M, McCardle T, Craig AG, Glembotski CC. Thecytoprotective effects of the glycoprotein 130 receptor-coupled cyto-kine, cardiotrophin-1, require activation of NF-kappa B. J Biol Chem2001;276:37621–9.

7. Shioi T, Kang PM, Douglas PS, et al. The conserved phosphoinositide3-kinase pathway determines heart size in mice. EMBO J 2000;19:2537–48.

8. Matsui T, Tao J, del Monte F, et al. Akt activation preserves cardiacfunction and prevents injury after transient cardiac ischemia in vivo.Circulation 2001;104:330–5.

9. Rosen BD, Edvardsen T, Lai S, et al. Left ventricular concentricremodeling is associated with decreased global and regional systolicfunction. The Multi-Ethnic Study of Atherosclerosis. Circulation2005;112:984–91.

0. Drazner MH. The transition from hypertrophy to failure: how certainare we? Circulation 2005;112:936–8.

1. Spinale FG, Coker ML, Bond BR, Zellner JL. Myocardial matrixdegradation and metalloproteinase activation in the failing heart: apotential therapeutic target. Cardiovasc Res 2000;46:225–38.

2. Cheng A, Nguyen TC, Malinowski M, et al. Effects of undersizedmitral annuloplasty on regional transmural left ventricular wallstrains and wall thickening mechanisms. Circulation 2006;114:I600 –9.

3. Wu AH, Aaronson KD, Bolling SF, Pagani FD, Welch K, KoellingTM. Impact of mitral valve annuloplasty on mortality risk in patientswith mitral regurgitation and left ventricular systolic dysfunction. J Am

Coll Cardiol 2005;45:381–7.