imaging survival and function of transplanted cardiac ... · bm-msc bone marrow-derived mesenchymal...

12
Imaging Survival and Function of Transplanted Cardiac Resident Stem Cells Zongjin Li, MD, PHD,* Andrew Lee, BS,* Mei Huang, PHD,* Hyung Chun, MD,† Jaehoon Chung, MD,† Pauline Chu, MS,§ Grant Hoyt, BS,‡ Phillip Yang, MD, PHD,† Jarrett Rosenberg, PHD,* Robert C. Robbins, MD,‡ Joseph C. Wu, MD, PHD*† Stanford, California Objectives The goal of this study is to characterize resident cardiac stem cells (CSCs) and investigate their therapeutic effi- cacy in myocardial infarction by molecular imaging methods. Background CSCs have been isolated and characterized in vitro. These cells offer a provocative method to regenerate the damaged myocardium. However, the survival kinetics and function of transplanted CSCs have not been fully elucidated. Methods CSCs were isolated from L2G85 transgenic mice (FVB strain background) that constitutively express both firefly luciferase and enhanced green fluorescence protein reporter gene. CSCs were characterized in vitro and trans- planted in vivo into murine infarction models. Multimodality noninvasive imaging techniques were used to as- sess CSC survival and therapeutic efficacy for restoration of cardiac function. Results CSCs can be isolated from L2G85 mice, and fluorescence-activated cell sorting analysis showed expression of resident CSC markers (Sca-1, c-Kit) and mesenchymal stem cell markers (CD90, CD106). Afterwards, 5 10 5 CSCs (n 30) or phosphate-buffered saline control (n 15) was injected into the hearts of syngeneic FVB mice undergoing left anterior descending artery ligation. Bioluminescence imaging showed poor donor cell survival by week 8. Echocardiogram, invasive hemodynamic pressure-volume analysis, positron emission tomography imag- ing with fluorine-18-fluorodeoxyglucose, and cardiac magnetic resonance imaging demonstrated no significant difference in cardiac contractility and viability between the CSC and control group. Finally, postmortem analysis confirmed transplanted CSCs integrated with host cardiomyocytes by immunohistology. Conclusions In a mouse myocardial infarction model, Sca-1–positive CSCs provide no long-term engraftment and benefit to cardiac function as determined by multimodality imaging. (J Am Coll Cardiol 2009;53:1229–40) © 2009 by the American College of Cardiology Foundation Recent years have brought stem cell therapy for heart disease to the forefront. Several clinical trials have shown beneficial effects of stem cell transplantation to improve cardiac function after myocardial infarction (MI) (1–3). However, the mechanism(s) behind this benefit is not clearly understood. Although numerous studies have sug- gested the cardioprotective benefits from various stem cell types, they also carry limitations including the potential of tumorigenicity with embryonic stem cells (4), arrhythmo- genicity with skeletal myoblasts (5), as well as existing controversies surrounding transdifferentiation of bone marrow-derived stem cells (6,7). The identification of undifferentiated cells with stem cell-like properties derived from the myocardium has re- newed the excitement in this field. A number of recent studies have confirmed the presence of cells bearing stem cell markers in the myocardium, which were successfully terminally differentiated into cell lineages that can benefit myocardial function (8 –12). A recent study even used resident cardiac stem cells (CSCs) derived from human myocardial biopsy specimens and injected them into mice, showing engraftment and improved myocardial function (12). We have previously described the L2G85 transgenic mice, which constitutively express both the firefly luciferase (Fluc) and enhanced green fluorescence protein (eGFP) genes (13,14). Cells derived from these mice provide the From the *Department of Radiology and Molecular Imaging Program (MIPS), †Department of Medicine, Division of Cardiology, ‡Department of Cardiothoracic Surgery, and the §Department of Comparative Medicine, Stanford University School of Medicine, Stanford, California. This work was supported, in part, by grants from the National Institutes of Health HL089027 (to Dr. Wu), the Burroughs Wellcome Foundation Career Award for Medical Scientists (to Dr. Wu), Stanford Cardiovas- cular Institute (to Dr. Wu), and the Society of Nuclear Medicine Pilot Research Award (to Dr. Li). Manuscript received September 26, 2008; revised manuscript received December 1, 2008, accepted December 23, 2008. Journal of the American College of Cardiology Vol. 53, No. 14, 2009 © 2009 by the American College of Cardiology Foundation ISSN 0735-1097/09/$36.00 Published by Elsevier Inc. doi:10.1016/j.jacc.2008.12.036

Upload: others

Post on 01-Nov-2019

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Imaging Survival and Function of Transplanted Cardiac ... · BM-MSC bone marrow-derived mesenchymal stem cell CSC cardiac stem cell(s) DAPI 4=6-diamidino-2-phenylindole DiI-ac-LDL

RdbcHcgtt

F†SotFcA

2

Journal of the American College of Cardiology Vol. 53, No. 14, 2009© 2009 by the American College of Cardiology Foundation ISSN 0735-1097/09/$36.00P

Imaging Survival and Function ofTransplanted Cardiac Resident Stem Cells

Zongjin Li, MD, PHD,* Andrew Lee, BS,* Mei Huang, PHD,* Hyung Chun, MD,†Jaehoon Chung, MD,† Pauline Chu, MS,§ Grant Hoyt, BS,‡ Phillip Yang, MD, PHD,†Jarrett Rosenberg, PHD,* Robert C. Robbins, MD,‡ Joseph C. Wu, MD, PHD*†

Stanford, California

Objectives The goal of this study is to characterize resident cardiac stem cells (CSCs) and investigate their therapeutic effi-cacy in myocardial infarction by molecular imaging methods.

Background CSCs have been isolated and characterized in vitro. These cells offer a provocative method to regenerate thedamaged myocardium. However, the survival kinetics and function of transplanted CSCs have not been fullyelucidated.

Methods CSCs were isolated from L2G85 transgenic mice (FVB strain background) that constitutively express both fireflyluciferase and enhanced green fluorescence protein reporter gene. CSCs were characterized in vitro and trans-planted in vivo into murine infarction models. Multimodality noninvasive imaging techniques were used to as-sess CSC survival and therapeutic efficacy for restoration of cardiac function.

Results CSCs can be isolated from L2G85 mice, and fluorescence-activated cell sorting analysis showed expression ofresident CSC markers (Sca-1, c-Kit) and mesenchymal stem cell markers (CD90, CD106). Afterwards, 5 � 105

CSCs (n � 30) or phosphate-buffered saline control (n � 15) was injected into the hearts of syngeneic FVB miceundergoing left anterior descending artery ligation. Bioluminescence imaging showed poor donor cell survival byweek 8. Echocardiogram, invasive hemodynamic pressure-volume analysis, positron emission tomography imag-ing with fluorine-18-fluorodeoxyglucose, and cardiac magnetic resonance imaging demonstrated no significantdifference in cardiac contractility and viability between the CSC and control group. Finally, postmortem analysisconfirmed transplanted CSCs integrated with host cardiomyocytes by immunohistology.

Conclusions In a mouse myocardial infarction model, Sca-1–positive CSCs provide no long-term engraftment and benefit tocardiac function as determined by multimodality imaging. (J Am Coll Cardiol 2009;53:1229–40) © 2009 bythe American College of Cardiology Foundation

ublished by Elsevier Inc. doi:10.1016/j.jacc.2008.12.036

gcm

cnsctmrmmf

m(

ecent years have brought stem cell therapy for heartisease to the forefront. Several clinical trials have showneneficial effects of stem cell transplantation to improveardiac function after myocardial infarction (MI) (1–3).owever, the mechanism(s) behind this benefit is not

learly understood. Although numerous studies have sug-ested the cardioprotective benefits from various stem cellypes, they also carry limitations including the potential ofumorigenicity with embryonic stem cells (4), arrhythmo-

rom the *Department of Radiology and Molecular Imaging Program (MIPS),Department of Medicine, Division of Cardiology, ‡Department of Cardiothoracicurgery, and the §Department of Comparative Medicine, Stanford University Schoolf Medicine, Stanford, California. This work was supported, in part, by grants fromhe National Institutes of Health HL089027 (to Dr. Wu), the Burroughs Wellcomeoundation Career Award for Medical Scientists (to Dr. Wu), Stanford Cardiovas-ular Institute (to Dr. Wu), and the Society of Nuclear Medicine Pilot Researchward (to Dr. Li).

gManuscript received September 26, 2008; revised manuscript received December 1,

008, accepted December 23, 2008.

enicity with skeletal myoblasts (5), as well as existingontroversies surrounding transdifferentiation of bonearrow-derived stem cells (6,7).The identification of undifferentiated cells with stem

ell-like properties derived from the myocardium has re-ewed the excitement in this field. A number of recenttudies have confirmed the presence of cells bearing stemell markers in the myocardium, which were successfullyerminally differentiated into cell lineages that can benefityocardial function (8–12). A recent study even used

esident cardiac stem cells (CSCs) derived from humanyocardial biopsy specimens and injected them intoice, showing engraftment and improved myocardial

unction (12).We have previously described the L2G85 transgenicice, which constitutively express both the firefly luciferase

Fluc) and enhanced green fluorescence protein (eGFP)

enes (13,14). Cells derived from these mice provide the
Page 2: Imaging Survival and Function of Transplanted Cardiac ... · BM-MSC bone marrow-derived mesenchymal stem cell CSC cardiac stem cell(s) DAPI 4=6-diamidino-2-phenylindole DiI-ac-LDL

tw01bpcwdtiCIc

fPawdcFisascBbaaamPLiasfImcIDteMSIs2wrmmCagDdfieepwawOsa

1230 Li et al. JACC Vol. 53, No. 14, 2009Transplantation of Cardiac Resident Stem Cells April 7, 2009:1229–40

advantage of being “trackable”over time. Using the previouslydescribed methods for CSC iso-lation and proliferation (10), wenow provide in detail the survivalkinetics of transplanted CSCsin infarcted murine hearts. Incontrast to previous studies(9,10,12,15,16), our results sug-gest that these CSCs do notconstitute long-term improve-ment in myocardial function. Wealso show definitive evidence viabioluminescence imaging (BLI)that the majority of these cellsare not viable by 8 weekspost-transplantation.

Methods

Isolation and culture of CSCs.Animal protocols were approvedby the Stanford University Ani-mal Care and Use Committee.The L2G85 transgenic mice ofFVB background with beta-actinpromoter driving Fluc-eGFPhave been described previously(14,17). Normal adult femaleFVB mice were used as recipientcontrols. CSCs were isolated from6- to 12-week-old L2G85 mice asdescribed with some modifications(10,11). In brief, myocardial tissuewas cut into a 1- to 2-mm piece,washed with Hanks’ balanced saltsolution (Invitrogen, Carlsbad,California), and incubated with0.1% collagenase II for 30 min at37°C with frequent shaking. Cellswere then filtered through 100-�mmesh. The cells obtained were cul-

ured in Iscove’s modified Dulbecco’s medium supplementedith 10% fetal bovine serum (FBS) (Hyclone, Logan, Utah),.1 mmol/l nonessential amino acids, 100 U/ml penicillin G,00 �g/ml streptomycin, 2 mmol/l glutamine, and 0.1 mmol/leta-mercaptoethanol. After 2 to 3 weeks, a population ofhase-bright cells appeared over the adhered fibroblast-likeells. These phase-bright cells were collected by 2 washesith phosphate-buffered saline (PBS), and 1 wash with cellissolution buffer (Gibco, Grand Island, New York) at roomemperature under microscopic monitoring, and subculturedn poly-lysine-coated plates (BD Pharmingen, San Jose,alifornia) with the same medium (Online Videos 1 and 2).

mmunofluorescence staining of cultured CSCs. The

Abbreviationsand Acronyms

BLI � bioluminescenceimaging

BM-MSC � bone marrow-derived mesenchymalstem cell

CSC � cardiac stem cell(s)

DAPI � 4=6-diamidino-2-phenylindole

DiI-ac-LDL � DiI acetylatedlow-density lipoprotein

EF � ejection fraction

eGFP � enhanced greenfluorescence protein

FBS � fetal bovine serum

Fluc � firefly luciferase

FS � fractional shortening

%ID/g � injected dose pergram of heart

LAD � left anteriordescending artery

LVEDd � left ventricularend-diastolic diameter

LVESd � left ventricularend-systolic diameter

MI � myocardial infarction

MRI � magnetic resonanceimaging

PBS � phosphate-bufferedsaline

PET � positron emissiontomography

PV � pressure-volume

ROI � region of interest

RT-PCR � reversetranscription-polymerasechain reaction

[18F]-FDG � fluorine-18-fluorodeoxyglucose

ells were fixed with 4% paraformaldehyde and processed a

or immunofluorescence with antibodies to Sca-1 (BDharmingen) in situ. Briefly, cells were incubated with ratntimouse Sca-1 for 1 h at room temperature and incubatedith goat antirat Alexa Fluor 594 (Invitrogen). 4=6-iamidino-2-phenylindole (DAPI) was used for nuclearounterstaining.low cytometry analysis. Fluorescence-activated cell sort-

ng (FACS) analysis of the bright cells and the cellsubcultured on poly-lysine-coated plates were carried out,nd the third passage of bone marrow-derived mesenchymaltem cells (BM-MSCs) from L2G85 mice was used asontrol cells. The isolation and culturing techniques ofM-MSCs were similar to a previous report (18). Anti-odies used in this study were phycoerythrin-conjugatednti-CD34, CD29, CD90, CD44, CD54, Flk1, Sca-1,llophycocyanin-conjugated anti-CD31 and c-Kit,llophycocyanin-conjugated antirat IgG2a, and rat anti-ouse CD62E, CD62P, Tie-2, CD144 (all from BDharmingen). The stained cells were analyzed using BDSR (BD Pharmingen). Dead cells stained by propidium-

odide were excluded from the analysis. Isotype-identicalntibodies served as controls (BD Pharmingen). FlowJooftware (Tree Star Inc., Ashland, Oregon) was used forollowed data analysis.n vitro differentiation of CSC. For cardiac and smoothuscle differentiation, CSCs were cultured in poly-lysine-

oated plates in differentiation medium containing 35%scove’s modified Dulbecco’s medium with 10% FBS/65%ulbecco’s modified Eagle medium-Ham F-12 mix con-

aining 2% B27, 0.1 mmol/l 2-mercaptoethanol, 10 ng/mlpidermal growth factor (R&D Systems Inc., Minneapolis,

innesota), 20 ng/ml basic fibroblast growth factor (R&Dystems Inc.), 40 nmol/l cardiotrophin-1 (R&D Systemsnc.), 40 nmol/l thrombin (Sigma-Aldrich, St. Louis, Mis-ouri), 100 U/ml penicillin G, 100 �g/ml streptomycin, andmmol/l glutamine (10). After 1 week, the cells were fixedith 4% paraformaldehyde and processed for immunofluo-

escence with antibodies specific to cardiac troponin T,yocyte enhancer factor 2C, connexin 43, and �-smoothuscle actin (all from Santa Cruz Biotech, Santa Cruz,alifornia) in situ. Briefly, cells were incubated with first

ntibody for 1 h at room temperature and incubated withoat antirabbit or donkey antimouse Alexa Fluor 594.API was used for nuclear counterstaining. For endothelial

ifferentiation, the phase-bright cells were cultured onbronectin-coated plates with EGM-2 (Cambrex, Walk-rsville, Maryland) with an extra 20 ng/ml of vascularndothelial growth factor. DiI acetylated low-density li-oprotein (DiI-ac-LDL) uptake assay and Matrigel assayere used to confirm endothelial cell phenotype differenti-

tion. For DiI-ac-LDL uptake assay, cells were incubatedith 10 �g/ml of DiI-ac-LDL (Molecular Probes, Eugene,regon) at 37°C for 6 h. After washing with PBS twice, the

lides were fixed and detected with fluorescence microscopys described (19,20). The formation of endothelial tubes was

ssessed by seeding cells in 24-well plates coated with
Page 3: Imaging Survival and Function of Transplanted Cardiac ... · BM-MSC bone marrow-derived mesenchymal stem cell CSC cardiac stem cell(s) DAPI 4=6-diamidino-2-phenylindole DiI-ac-LDL

MftFmi�ebcplbd3osdMfRetiipawJwt1i�PawCwc(teauOf5Ct5Dmwms

C(iHfPoi[aIp(I(dmtFasmmaALawaobSs12LscmtwCmeisscIaMIAn

1231JACC Vol. 53, No. 14, 2009 Li et al.April 7, 2009:1229–40 Transplantation of Cardiac Resident Stem Cells

atrigel (BD Pharmingen) and incubating them at 37°Cor 12 h as described (19,20). For adipogenic differentiation,he phase-bright cells were cultured in DMEM with 10%BS, 100 U/ml penicillin G, 100 �g/ml streptomycin, 2mol/l glutamine, 1 �mol/l dexamethasone, 10 �g/ml

nsulin, 0.5 mmol/l isobulyl-1-methylxanthione, and 200mol/l indomethacin for 2 weeks and medium was changedvery 3 days. The differentiated adipocytes were confirmedy oil red staining and reverse transcription-polymerasehain reaction (RT-PCR) analysis for peroxisomeroliferator-activated receptors gamma and lipoprotein

ipase expression. For osteogenic differentiation, the phase-right cells were cultured in osteogenic differentiation me-ium (Cambrex, Walkersville, Maryland; PT-3002) for 2 to

weeks and medium was changed every 3 days. Thesteocyte differentiation was confirmed by Alizarin red Staining and RT-PCR analysis for osteocalcin expression asescribed (21).

I and cell delivery. All surgical procedures were per-ormed on 8- to 10-week-old female FVB mice (Charlesiver Laboratories, Inc., Davis, California) by a single

xperienced micro-surgeon (G.H.). FVB mice were anes-hetized with inhaled isoflurane (2% to 3%). Mice werentubated, ventilated, and anesthesia was maintained withnhaled isoflurane (1% to 2.5%). A left thoracotomy waserformed, and the pericardium was opened. The leftnterior descending artery (LAD) was permanently ligatedith a 9-0 Ethilon suture (Ethicon Inc., Somerville, New

ersey) just distal to the level of the left atrium. Infarctionas visually confirmed by blanching of the anterior region of

he left ventricle along with dyskinesis. After 30 min, 5 �05 CSCs were injected intramyocardially into the peri-nfarct zone at 2 different sites with a total volume of 20l in each animal (n � 30). Control animals received aBS injection instead (n � 15). For sham-operatednimals (n � 5), open thoracotomy was performedithout suturing of the LAD and without injection ofSCs or PBS. A thoracotomy tube was placed and lungsere re-expanded using positive pressure ventilation. The

hest cavity was closed in layers with 5-0 Vicryl sutureEthicon Inc.), and the animal was gradually weaned fromhe respirator. Once spontaneous respiration resumed, thendotracheal and thoracotomy tubes were removed, and thenimals were placed in a temperature-controlled chamberntil they resumed full alertness and mobility.ptical BLI of transplanted cell survival. BLI was per-

ormed on all animals (n � 30 CSC, n � 15 PBS, and n �sham) using the IVIS 200 system (Xenogen, Alameda,alifornia) by a blinded investigator (M.H.). A majority of

he animals were scanned on days 2, 7, 14, 21, 28, 49, and6. After intraperitoneal injection of the reporter probe-Luciferin (150 mg/kg), animals were imaged for 1 to 10in. The same mice were imaged longitudinally for up to 8eeks. Bioluminescence signals were quantified in units ofaximum photons per second per centimeter square per

teridian (photons/s/cm2/sr) as described (19). M

ardiac viability with fluorine-18-fluorodeoxyglucose[18F]-FDG) positron emission tomography (PET) imag-ng. Small animal microPET imaging (Vista system, GE

ealthcare, Chalfont St. Giles, United Kingdom) was per-ormed in a subset of the animals (n � 14 CSC and n � 8BS) on baseline (day �7) and days 2, 28, and 56 post-peratively. Mice were fasted for 3 h before radioisotopenjection. Animals were then injected with 139 � 23 �Ci18F]-FDG via the tail vein. At 45 to 60 min after injection,nimals were anesthetized with inhaled 2% isoflurane.mages were performed, reconstructed by filtered backrojection, and analyzed using image software AmideAmide’s Medical Image Data Examiner, SourceForge,nc., Mountain View, California) by a blinded investigatorA.L.). Three-dimensional regions of interest (ROIs) wererawn encompassing the heart. For each ROI, counts/ml/in were then converted to counts/g/min and divided by

he injected dose to obtain the image ROI derived [18F]-DG percentage injected dose per gram of heart (% ID/g)s described (22). To measure the myocardium infarctionize, [18F]-FDG PET images were assembled into polaraps (23). The size of perfusion defect was measured as theyocardium with 50% of maximum activity and expressed

s percent total myocardium polar map by GE Healthcaredvantage Workstation.eft ventricular functional analysis with echocardiogram

nd pressure-volume (PV) acquisition. Echocardiographyas performed in all animals (n � 30 CSC, n � 15 PBS,

nd n � 5 sham). A majority of the animals were scannedn baseline (day �7) and days 2, 28, and 56 post-operativelyy a blinded investigator (M.H.) using the Siemens-Acusonequoia C512 system (Siemens Healthcare, Malvern, Penn-ylvania) equipped with a multifrequency (8 to 14 MHz)5L8 transducer. Animals were anesthetized with inhaled% isoflurane. Analysis of M-mode images was performed.eft ventricular end-diastolic diameter (LVEDd) and end-

ystolic diameter (LVESd) were measured and used toalculate fractional shortening (FS) by the following for-ula: FS � [LVEDd � LVESd]/LVEDd. At the end of

he study (week 8), invasive hemodynamic measurementsere performed in a subset of the animals (n � 5/group forSC, PBS, and sham) as described (24). Briefly, afteridline neck incision, a conductance 1.4 conductance cath-

ter (Millar Instruments, Houston, Texas) was introducednto the left ventricle through the right carotid artery. Aftertabilization, the signals were continuously recorded at aampling rate of 1,000/s using PV conductance systemoupled to a PowerLab/4SP analog to digital converter (ADnstruments, Colorado Springs, Colorado). Data were an-lyzed by using a cardiac PV analysis program (PVAN 3.4,

illar Instruments) and Chart/Scope Software (ADnstruments).ssessment of cardiac contractility with magnetic reso-ance imaging (MRI). Please see the Online Supplemental

ethods section.
Page 4: Imaging Survival and Function of Transplanted Cardiac ... · BM-MSC bone marrow-derived mesenchymal stem cell CSC cardiac stem cell(s) DAPI 4=6-diamidino-2-phenylindole DiI-ac-LDL

Htincnsdat5p

daccudf4fisIa

1232 Li et al. JACC Vol. 53, No. 14, 2009Transplantation of Cardiac Resident Stem Cells April 7, 2009:1229–40

istological analysis. After completion of the imaging pro-ocols, all animals were sacrificed at week 8. For immunostain-ng, explanted hearts (n � 10 for CSC, n � 5 for PBS, and

� 2 for sham) were embedded into Tissue-Tek O.C.T.ompound (Sakura Finetek USA Inc., Torrance, Califor-ia), snap frozen in liquid nitrogen, and cut into transverseections at 5-�m thickness. To track GFP-positive CSC-erived cells and cardiomyocytes differentiation in hearts,nti-GFP antibody (Invitrogen) and anti-�-sarcomeric ac-in antibody (Sigma) were used. Alexa Fluor 488- and Fluor94-conjugated secondary antibodies were applied appro-riately. DAPI was used for nuclear counterstaining. To

Figure 1 Isolation and Culturing of CSCs

(A) Ventricular cells from L2G85 transgenic mice stained for Sca-1 (red) (I). After a pcells (II). The phase-bright cells were collected, and Giemsa stain showed the cellin vitro differentiation of phase-bright cells. Morphology of cardiac stem cells (CSC(II). With cardiac differentiation medium, some CSCs can form cardiac sphere (arrlinear growth of cultured CSCs over a 1-month period. (D and E) Ex vivo imaging a

0.98). Also see Online Videos 1 and 2.

etect microvascular density in the peri-infarct area, a ratntimouse CD31 (BD Pharmingen) and Alexa Fluor 647-onjugated secondary antibody were used. The number ofapillary vessels was counted in 10 randomly selected areassing a fluorescence microscope (200� magnification). Toetect fibrosis in cardiac muscles, explanted hearts (n � 10or CSC, n � 5 for PBS, and n � 2 for sham) were fixed in% paraformaldehyde, cut transversely, embedded in paraf-n, and stained with Masson’s trichome (25). Five to 6ections from apex to base were subjected to staining.mages of each section were taken to calculate the fibroticnd nonfibrotic areas as well as ventricular and septal wall

nging from 1 to 3 weeks, phase-bright cells migrated over a layer of fibroblast-likelarge nucleus (III). Scale bar � 50 �m (I, II), � 2 �m (III). (B) Subculture andultured in poly-D-lysine-coated plates and expressing green fluorescence proteinith green fluorescence protein expression (III, IV). (C) Proliferation curves showof CSCs shows increasing bioluminescence signals with cell numbers (r2 �

eriod rawith a

s) (I) cow) wnalysis

Page 5: Imaging Survival and Function of Transplanted Cardiac ... · BM-MSC bone marrow-derived mesenchymal stem cell CSC cardiac stem cell(s) DAPI 4=6-diamidino-2-phenylindole DiI-ac-LDL

tmBtS(argcsamfsmdS

sw

R

Isotwh(psob(f

1233JACC Vol. 53, No. 14, 2009 Li et al.April 7, 2009:1229–40 Transplantation of Cardiac Resident Stem Cells

hickness. The measurement of area of fibrosis was deter-ined by Image J software (National Institutes of Health,ethesda, Maryland) performed on 3 separate sections, and

he averages were used for statistical analysis.tatistical analysis. Comparison of FS, ejection fractionEF), cardiac viability, and infarction size between CSCnd sham/PBS groups over time was done by 2-wayepeated measures analysis of variance (ANOVA) withroup and day as factors, using a Greenhouse-Geisserorrection for lack of sphericity. Comparison of end-ystolic and -diastolic volume and pressure between CSCnd sham/PBS groups was done by 2-way repeatedeasures ANOVA with group and cardiac phase as

actors, using a Greenhouse-Geisser correction for lack ofphericity. Comparison of histological infarct size andicrovascular density between PBS and CSC groups was

one by 2-tailed t test. Statistics were calculated usingPSS 14.0 (SPSS Inc., Chicago, Illinois). Descriptive

Figure 2 Characterization of Surface Markers in CSCs

Quantification by FACS analysis of cardiac stem cells (CSCs), phase-bright cells, a(BM-MSCs) were used as a control. (A) CSCs express robust green fluorescence pthere was up-regulation of Sca-1. Left panels show Sca-1 antibody isotype control.mesenchymal stem cells markers, CD29, CD90, CD44, and CD106. After subcultulated. Compared with BM-MSCs, CSCs express less CD54, but higher CD29 and C

tatistics included mean and standard error. Differencesere considered significant at p values of �0.05.

esults

solation and culturing of CSCs. Explanted hearts wereubjected to enzymatic digestion, cultured, and are composedf Sca-1–positive cells (Fig. 1A). Phase bright spherical cellshat spontaneously separated from the myocardium samplesere identified after 2 to 3 weeks. These cells demonstrated aigh nucleus-to-cytoplasm ratio and expressed robust GFPFig. 1B). The cells also demonstrated linear growth withopulation doubling time of approximately 5 days (Fig. 1C),imilar to previous studies (10,12,16). Importantly, assessmentf Fluc expression showed a linear correlation (r2 � 0.98)etween the total cell numbers and the bioluminescence signalsFigs. 1D and 1E), which is needed for accurate tracking of cellate in subsequent in vivo imaging studies.

ir derived cells. Bone marrow mononuclear cell-derived mesenchymal stem cells(GFP) both on phase-bright CSCs and their subcultures. After 2 to 3 passages,uantitative analysis of cell markers expression by FACS. CSCs express high-levelca-1 up-regulated, but c-Kit, CD34, CD31, and CD144 markers all down-regu-

All fluorescence-activated cell sorting experiments were performed in triplicate.

nd therotein(B) Qring, SD90.

Page 6: Imaging Survival and Function of Transplanted Cardiac ... · BM-MSC bone marrow-derived mesenchymal stem cell CSC cardiac stem cell(s) DAPI 4=6-diamidino-2-phenylindole DiI-ac-LDL

CsprwS(pOmCmciaFsfOsMt

ocpm(dsntcasasIgvhcqlC

1234 Li et al. JACC Vol. 53, No. 14, 2009Transplantation of Cardiac Resident Stem Cells April 7, 2009:1229–40

haracterization of surface markers in CSCs. Previoustudies have shown that Sca-1–expressing cells possess theotential to differentiate into cardiomyocytes (10,11). Ouresults showed that �40.0 � 5.5% of the phase-bright cellsere Sca-1 positive. After subculturing for 1 to 2 weeks,ca-1 expression on CSCs was up-regulated close to 95%Fig. 2A). In order to further characterize their cellularhenotypes, detailed flow cytometry analysis was performed.verall, the subcultured group had higher expression ofesenchymal stem cell markers (CD29, CD90, CD44,D106), but endothelial (CD31, CD34, CD144) and c-Kitarkers were both down-regulated (Fig. 2B). Interestingly,

ompared with BM-MSCs, subcultured CSCs have a sim-lar expression pattern, but express higher CD29 and CD90nd lower CD54. CSCs isolated from normal syngeneicVB mice also showed similar results (data not shown),uggesting no significant effects on cellular characteristicsrom the stable expression of the Fluc-eGFP reporter gene.

verall, the expression of these markers in our CSCs isimilar to previously published data (12,26).

ultipotent differentiation of CSCs. We next examinedhe differentiation of CSCs into various cell types. Similar to

Figure 3 Multipotent Capacity of CSCs

(A) Cardiac and smooth muscle differentiation of cardiac stem cells (CSCs) in vitr(cTnT), myocyte enhancer factor 2C (MEF-2C), connexin-43, and �-smooth muscleEGM-2 medium with 10 ng/ml vascular endothelial growth factor and showed endolipoprotein (II). Endothelial tube formation by differentiated CSCs after 12 h of platures (IV). (C) Oil red staining and reverse transcription-polymerase chain reactionlipase (LPL) expression shows adipogenic differentiation of the CSCs induced for 2chain reaction analysis of osteocalcin expression show CSCs induced to differenti

ther studies (10,21,27,28), these cells can differentiate intoardiomyocytes and smooth muscle cells, as documented byositive staining for cardiac troponin T, connexin 43,yocyte enhancer factor 2C, and �-smooth muscle actin

Fig. 3A). After 7 days in differentiation medium, the cellseveloped into clusters, and after 2 weeks they formed intopherical collection of cells, some of which showed sponta-eous contractility (Online Videos 1 and 2). In addition,hese cells were capable of differentiating into endothelialells as demonstrated by DiI-ac-LDL uptake and Matrigelngiogenesis assays (Fig. 3B), into adipocytes as demon-trated by oil red staining and RT-PCR analysis (Fig. 3C),nd into osteoblasts as demonstrated by Alizarin red Staining and RT-PCR analysis (Fig. 3D).njection of CSCs into infarcted myocardium. Severalroups have reported that CSCs can proliferate indefinitely initro (10,21,27,28). This begs the question whether CSCs canave similar growth properties in vivo, and if so, whether thisould lead to potential tumorigenesis. In order to address thisuestion, adult FVB mice were subjected to LAD arteryigation followed by injection with either 5 � 105 culturedSCs (n � 30) or PBS (n � 15). Injection of CSCs into

unostaining of green fluorescence protein-positive CSCs with cardiac troponin TSMA). (B) Endothelial differentiation of CSCs in vitro. The cells were cultured inl differentiation by morphology (I) and uptake of DiI acetylated low-density

Matrigel (III) whereas undifferentiated CSCs cells do not form cord-like struc-sis of peroxisome proliferator-activated receptors gamma (PPAR�) and lipoproteins. (D) Alizarin red S staining of calcium and reverse transcription-polymeraseo osteoblasts.

o. Immactin (theliating onanalyweek

ate int

Page 7: Imaging Survival and Function of Transplanted Cardiac ... · BM-MSC bone marrow-derived mesenchymal stem cell CSC cardiac stem cell(s) DAPI 4=6-diamidino-2-phenylindole DiI-ac-LDL

icsdrwa(iAsd(gptgb2tcssF

scSvsF2pCAaataav2staart

1235JACC Vol. 53, No. 14, 2009 Li et al.April 7, 2009:1229–40 Transplantation of Cardiac Resident Stem Cells

nfarcted myocardium resulted in a robust biolumines-ence signal at day 2. However, serial imaging of theame animals out to 8 weeks demonstrated a significantecay in the bioluminescence signal. When normalized toesults from day 2, the percent bioluminescence signalsere 6.7 � 0.5 at day 7, 3.9 � 0.8 at day 14, 1.7 � 0.2

t day 21, 1.4 � 0.1 at day 28, and 0.4 � 0.1 at day 56Fig. 4). Control animals injected with PBS showed nomaging signals as expected.

ssessment of cardiac contractility and hemodynamictatus after CSC therapy. To date, only a few studies haveemonstrated that CSCs can improve cardiac function10,21,27,28). In order to confirm these results, echocardio-rams were performed at baseline, day 2, day 28, and day 56ost-surgery (Figs. 5A and 5B). At day 2 after infarction,here was a significant decrease in the FS in both the CSCroup and the PBS group compared with that seen inaseline, consistent with successful induction of MI. At days8 and 56, there was a trend toward slight improvement inhe FS for both groups, but the difference was not statisti-ally significant. These results were also confirmed in aubset of the animals that underwent cardiac MRI, whichhowed no functional change in EF (Online Supplemental

Figure 4 Reporter Gene Imaging of CSC Fate After Transplanta

(A) A representative animal injected with 5 � 105 cardiac stem cells (CSCs) showfollowing 8 weeks. (B) Detailed quantitative analysis of signals from all animals trtion of percent donor cell survival plotted as % signal activity (normalized to day 2

ig. 1). In addition, invasive hemodynamic parameters P

howed a decrease in EF for both CSC and control groupsompared with that seen in the sham group (Onlineupplemental Fig. 2). This was associated with higher leftentricular end-diastolic volume and left ventricular end-ystolic volume in both groups compared with sham.inally, as highlighted in the Online Supplemental Table, there was no significant difference in other functionalarameters (e.g., cardiac output, stroke volume) betweenSC transplanted mice and PBS control mice.ssessment of cardiac viability after CSC therapy. In

ddition, we performed [18F]-FDG PET scans of the micet baseline, day 2, day 28, and day 56 post-surgery. Thisechnique can be used to detect changes in cardiac viabilityfter interventions in small animal models (29). At day 2fter infarction, there was a significant reduction in cardiaciability for both groups (20.2 � 4.9% ID/g for PBS and0.8 � 4.2% ID/g for CSCs; p � 0.001), consistent withuccessful induction of MI. However, at day 28 and day 56,here was no significant difference between the treatmentrm and the control group (Figs. 6A and 6B). Moreover,nalysis of the [18F]-FDG PET images using polar-mapeconstruction revealed no significant difference of infarc-ion size between the CSC transplantation group and the

ificant bioluminescence activity at day 2, which decreases progressively over thented with CSCs. Signal activity is expressed as photons/s/cm2/sr. (C) Estima-the 8-week period after transplantation. PBS � phosphate-buffered saline.

tion

s signanspla) over

BS group (Figs. 6C and 6D).

Page 8: Imaging Survival and Function of Transplanted Cardiac ... · BM-MSC bone marrow-derived mesenchymal stem cell CSC cardiac stem cell(s) DAPI 4=6-diamidino-2-phenylindole DiI-ac-LDL

HmfaswCfissddthaS

D

ILeidSmi

mhawmimcMt

v(licattimdc�do

1236 Li et al. JACC Vol. 53, No. 14, 2009Transplantation of Cardiac Resident Stem Cells April 7, 2009:1229–40

istological assessment of infarct size and CSC engraft-ent. Histological analysis of the myocardium was per-

ormed by both looking at thin sections of the gross specimennd via immunofluorescent microscopic examination. Micro-copic examination showed the presence of GFP-positive cellsithin myocardium at day 3 (Figs. 7A and 7B). At day 14,SCs could differentiate into cardiomyocytes as con-rmed by �-smooth muscle actin and GFP doubletainings (Fig. 7C). However, this population becameignificantly decreased when the tissues were examined atay 28 (Fig. 7D), which is also consistent with theecrease in bioluminescence signals over this period ofime (Fig. 4). Finally, examination of the explantedearts showed no significant difference in the infarct sizend microvascular density between the 2 groups (Onlineupplemental Fig. 3).

iscussion

n this study, we isolated cardiac resident stem cells from2G85 transgenic mice that constitutively expressed Fluc-GFP, enabling us to track stem cells by both noninvasivemaging and invasive histopathology. Upon culturing, weemonstrated that these phase-bright cells up-regulatedca-1 and expressed surface markers resembling mesenchy-al stem cells. We also present evidence that molecular

Figure 5 Echocardiographic Evaluation of Cardiac Contractility

(A) Representative M-mode echocardiographic data of infarcted hearts receiving Pshortening (FS) and ejection fraction (EF) between the 2 groups 7 days before (basligation. Abbreviations as in Figure 4.

maging can be used to track CSCs survival in a murine v

odel of MI. When injected directly into the infarctedeart, these cells initially demonstrated viability by BLI. Butfter a period of 8 weeks, the majority of the imaging signalsere no longer present, suggesting elimination from theyocardium. Consequently, we found no significant phys-

ologic benefit from CSC therapy as determined fromultiple parameters, including FS by echocardiogram, myo-

ardial glucose uptake by [18F]-FDG PET scan, EF byRI, invasive PV loop analysis, and infarct size by his-

opathological assessment.Recent works have documented the presence of a reser-

oir of stem and progenitor cells in the myocardium8,10,16,21,30–32). These cells have been successfully iso-ated and expanded ex vivo, and have been differentiatednto cardiomyocytes, smooth muscle cells, and endothelialells (10,16,21,26,30–32). Previous works using CSCs havelso demonstrated significant improvement in cardiac func-ion after CSC injection, which was, in part, attributed tohe persistent engraftment of the injected CSCs into thenfarct zone (9,12,16,33). A higher percentage of viable

yocardium within the infarct zone and improved EF wereemonstrated up to 5 weeks after cell transplantation (9). Inontrast, BLI data from our study suggest that by week 8,0.5% of the transplanted CSCs are still alive. By echocar-

iography and PET imaging, no significant changes werebserved at day 28 and day 56 in cardiac contractility and

sus CSCs at day 2, week 4, and week 8. (B and C) Comparison of fractional, 2 days, 28 days, and 56 days after left anterior descending coronary artery

BS vereline)

iability.

Page 9: Imaging Survival and Function of Transplanted Cardiac ... · BM-MSC bone marrow-derived mesenchymal stem cell CSC cardiac stem cell(s) DAPI 4=6-diamidino-2-phenylindole DiI-ac-LDL

finep(Fpio

csgsssmiB

1237JACC Vol. 53, No. 14, 2009 Li et al.April 7, 2009:1229–40 Transplantation of Cardiac Resident Stem Cells

At present, we can only speculate that the discrepancy inndings may be attributed to differences in isolation tech-iques (cardiosphere, side population), stem cell markerxpression (c-Kit, Sca-1), donor cell sources (human, mice),articular strains of mice used, or host animal modelsnonreperfused LAD ligation, ischemia-reperfusion) (34).rom the procedural standpoint, we cannot exclude that theossibility that our mice may have been oversedated (2%soflurane) given the lower baseline FS values (35% to 40%)

Figure 6 [18F]-FDG PET Imaging of Cardiac Viability

(A) Representative image in a normal mouse heart and at days 2, 28, and 56 in igroup at days 28 and 56, respectively. (B) Detailed quantitative analysis of signalence of the FDG uptake (injected dose per gram of heart [% ID/g]) between the 2(C and D) Representative polar map of the micropositron emission tomography (Pbased on 50% thresholds. [18F]-FDG � fluorine-18-fluorodeoxyglucose; other abbre

bserved compared with those in previous studies (35). This (

ould have undermined the chances of detecting a small butignificant change between the 2 groups. From the trans-enic model standpoint, it is possible that Fluc reporter geneilencing may have occurred and limited our ability to detecturviving CSCs at late time points. However, a previoustudy using hematopoietic stem cells isolated from theseice was able to demonstrate complete reconstitution of the

rradiated bone marrow at late time points as detected byLI, which argues against significant transgene silencing

d hearts receiving PBS versus CSCs. *p � 0.46 and p � 0.38 versus PBSall animals transplanted with CSCs and PBS group. There is no significant differ-. *p � 0.31 and p � 0.40 versus PBS group at days 28 and 56, respectively.

ages obtained from mice treated with PBS versus CSC. Measurements ares as in Figure 4.

nfarctes fromgroupsET) imviation

17). From the technical standpoint, BLI is limited to small

Page 10: Imaging Survival and Function of Transplanted Cardiac ... · BM-MSC bone marrow-derived mesenchymal stem cell CSC cardiac stem cell(s) DAPI 4=6-diamidino-2-phenylindole DiI-ac-LDL

abpActidasbnv

mmp

sptc(oiamtbettmt

1238 Li et al. JACC Vol. 53, No. 14, 2009Transplantation of Cardiac Resident Stem Cells April 7, 2009:1229–40

nimal imaging as the low-energy photons (2 to 3 eV) canecome attenuated within deeper tissues (e.g., heart) com-ared with more superficial locations (e.g., skeletal muscles).t present, a conservative estimate of minimal detectable

ell number by BLI is �1,000 in the heart versus �100 inhe leg (14). Finally, from the instrumentation standpoint, its plausible that the image resolution (�1.5 mm3) andetection sensitivity (10�9 to 10�12 mol/l) of the smallnimal microPET scanner is still insufficient to distinguishubtle but significant differences in [18F]-FDG uptakeetween the 2 groups. Thus, additional studies will beeeded in the future to further examine each of theseariables separately.

Just as the mechanism of stem cells exerting benefit onyocardial function remains to be fully elucidated, theechanism of their elimination over time also remains

Figure 7 Tracking of Grafted CSCs by Immunofluorescence

(A and B) Cardiac stem cells (CSCs) within the recipient myocardium 3 days afterferentiate and integrate with host myocardium as confirmed by green fluorescenceferentiate into cardiomyocytes as confirmed by �-SA and GFP double stainings (C)examined at day 28 (D), which is also consistent with the decrease in biolumines

oorly understood. The pattern of acute donor cell death t

een in our CSC transplantation is also consistent withrevious studies showing poor donor cell survival afterransplantation of neonatal cardiomyocytes (36), mesen-hymal stem cells (37), bone marrow mononuclear cells14), and human embryonic stem cell-derived cardiomy-cytes (38). A number of culprits may be involved,ncluding inflammation, ischemia, apoptosis, anoikis, andutophagy, and may all play contributory roles. Further-ore, the myocardial milieu in vivo is in stark contrast to

he rich nutrients and oxygen concentration that haveeen optimized for their cell culturing, growth, andxpansions in vitro. It is interesting to note historicallyhat myoblast transplantation as a potential cell-basedherapy for patients with Duchenne muscular dystrophyet with disappointing failures in the 1990s (39). One of

he main reasons was acute donor cell death after transplanta-

on shown at low and high magnification. (C and D) Transplanted CSCs can dif-in (GFP) and �-sarcomeric actin (SA) double staining. At day 14, CSCs could dif-ver, this population became significantly decreased when the tissues weresignals over this period of time. Scale bar � 100 �m (A), 20 �m (B to D).

injectiprote

. Howecence

ion. Future clinical studies aimed at transplanting CSCs in

Page 11: Imaging Survival and Function of Transplanted Cardiac ... · BM-MSC bone marrow-derived mesenchymal stem cell CSC cardiac stem cell(s) DAPI 4=6-diamidino-2-phenylindole DiI-ac-LDL

pa

bepspfnmpeTibia

C

ItCfcuirfn

RSS

R

1

1

1

1

1

1

1

1

1

1

2

2

2

2

2

2

2

2

2

2

3

3

3

3

1239JACC Vol. 53, No. 14, 2009 Li et al.April 7, 2009:1229–40 Transplantation of Cardiac Resident Stem Cells

atients will likely need to first understand the mechanism ofcute donor cell death.

Despite the limitation of acute donor stem cell death,oth animal and clinical studies have suggested beneficialffects after transplantation of various stem cell types (40),erhaps acting through paracrine pathways (41). Thus,trategies aimed at prolonging cell survival (and, hence,rolonging paracrine activation) may lead to even moreavorable results. To that extent, the application of bioengi-eering methods (42), pro-survival cocktails (43), or geneticodification (44), rather than direct stem cell injection, may

rove to be a more viable approach for achieving long-termngraftment in the future. Encouragingly, a recent study byillmanns et al. (45) showed that CSCs activated with

nsulin-like growth factor 1 and hepatocyte growth factorefore their injection into infarcted sites had significantlymproved cell survival rate and were able to form conductiverterioles and capillaries in the host myocardium.

onclusions

n summary, our study demonstrates that imaging can be usedo monitor CSC fate noninvasively in living animals and thatSC transplantation provides no significant benefits to cardiac

unction in a mouse MI model. As the field of CSC therapyontinues to mature, it is critical to better understand thenderlying mechanism of the treatment modality, and todentify the potential pitfalls of the therapy before its incorpo-ation into the clinical realm (1). Although a stem cell sourcerom the heart is of tremendous potential, cautious optimism isecessary before full clinical use of these cells.

eprint requests and correspondence: Dr. Joseph C. Wu,tanford University School of Medicine, Edwards Building, R354,tanford, California 94305-5344. E-mail: [email protected].

EFERENCES

1. Rosenzweig A. Cardiac cell therapy—mixed results from mixed cells.N Engl J Med 2006;355:1274–7.

2. Assmus B, Honold J, Schachinger V, et al. Transcoronary transplan-tation of progenitor cells after myocardial infarction. N Engl J Med2006;355:1222–32.

3. Wollert KC, Meyer GP, Lotz J, et al. Intracoronary autologousbone-marrow cell transfer after myocardial infarction: the BOOSTrandomised controlled clinical trial. Lancet 2004;364:141–8.

4. Cao F, Lin S, Xie X, et al. In vivo visualization of embryonic stem cellsurvival, proliferation, and migration after cardiac delivery. Circulation2006;113:1005–14.

5. Menasche P, Alfieri O, Janssens S, et al. The Myoblast AutologousGrafting in Ischemic Cardiomyopathy (MAGIC) trial: first random-ized placebo-controlled study of myoblast transplantation. Circulation2008;117:1189–200.

6. Balsam LB, Wagers AJ, Christensen JL, Kofidis T, Weissman IL,Robbins RC. Haematopoietic stem cells adopt mature haematopoieticfates in ischaemic myocardium. Nature 2004;428:668–73.

7. Orlic D, Kajstura J, Chimenti S, et al. Bone marrow cells regenerateinfarcted myocardium. Nature 2001;410:701–5.

8. Beltrami AP, Barlucchi L, Torella D, et al. Adult cardiac stem cellsare multipotent and support myocardial regeneration. Cell 2003;114:763–76.

9. Dawn B, Stein AB, Urbanek K, et al. Cardiac stem cells deliveredintravascularly traverse the vessel barrier, regenerate infarcted myocar-

dium, and improve cardiac function. Proc Natl Acad Sci U S A2005;102:3766–71.

0. Messina E, De Angelis L, Frati G, et al. Isolation and expansion ofadult cardiac stem cells from human and murine heart. Circ Res2004;95:911–21.

1. Oh H, Bradfute SB, Gallardo TD, et al. Cardiac progenitor cells fromadult myocardium: homing, differentiation, and fusion after infarction.Proc Natl Acad Sci U S A 2003;100:12313–8.

2. Smith RR, Barile L, Cho HC, et al. Regenerative potential ofcardiosphere-derived cells expanded from percutaneous endomyocar-dial biopsy specimens. Circulation 2007;115:896–908.

3. van der Bogt KE, Sheikh AY, Schrepfer S, et al. Comparison ofdifferent adult stem cell types for treatment of myocardial ischemia.Circulation 2008;118:S121–9.

4. Sheikh AY, Lin SA, Cao F, et al. Molecular imaging of bone marrowmononuclear cell homing and engraftment in ischemic myocardium.Stem Cells 2007;25:2677–84.

5. Gonzalez A, Rota M, Nurzynska D, et al. Activation of cardiacprogenitor cells reverses the failing heart senescent phenotype andprolongs lifespan. Circ Res 2008;102:597–606.

6. Ott HC, Matthiesen TS, Brechtken J, et al. The adult human heart asa source for stem cells: repair strategies with embryonic-like progenitorcells. Nat Clin Pract Cardiovasc Med 2007;4 Suppl 1:S27–39.

7. Cao YA, Wagers AJ, Beilhack A, et al. Shifting foci of hematopoiesisduring reconstitution from single stem cells. Proc Natl Acad Sci U S A2004;101:221–6.

8. Sun S, Guo Z, Xiao X, et al. Isolation of mouse marrow mesen-chymal progenitors by a novel and reliable method. Stem Cells2003;21:527–35.

9. Li Z, Wu JC, Sheikh AY, et al. Differentiation, survival, and functionof embryonic stem cell derived endothelial cells for ischemic heartdisease. Circulation 2007;116:I46–54.

0. Li ZJ, Wang ZZ, Zheng YZ, et al. Kinetic expression of plateletendothelial cell adhesion molecule-1 (PECAM-1/CD31) during em-bryonic stem cell differentiation. J Cell Biochem 2005;95:559–70.

1. Matsuura K, Nagai T, Nishigaki N, et al. Adult cardiac Sca-1–positivecells differentiate into beating cardiomyocytes. J Biol Chem 2004;279:11384–91.

2. Toyama H, Ichise M, Liow JS, et al. Evaluation of anesthesia effectson [18F]FDG uptake in mouse brain and heart using small animalPET. Nucl Med Biol 2004;31:251–6.

3. Kudo T, Fukuchi K, Annala AJ, et al. Noninvasive measurement ofmyocardial activity concentrations and perfusion defect sizes in ratswith a new small-animal positron emission tomograph. Circulation2002;106:118–23.

4. Xu M, Uemura R, Dai Y, Wang Y, Pasha Z, Ashraf M. In vitro andin vivo effects of bone marrow stem cells on cardiac structure andfunction. J Mol Cell Cardiol 2007;42:441–8.

5. Engel FB, Hsieh PC, Lee RT, Keating MT. FGF1/p38 MAP kinaseinhibitor therapy induces cardiomyocyte mitosis, reduces scarring, andrescues function after myocardial infarction. Proc Natl Acad Sci U S A2006;103:15546–51.

6. Tateishi K, Ashihara E, Honsho S, et al. Human cardiac stem cellsexhibit mesenchymal features and are maintained through Akt/GSK-3beta signaling. Biochem Biophys Res Commun 2007;352:635–41.

7. Barile L, Chimenti I, Gaetani R, et al. Cardiac stem cells: isolation,expansion and experimental use for myocardial regeneration. Nat ClinPract Cardiovasc Med 2007;4 Suppl 1:S9–14.

8. Bearzi C, Rota M, Hosoda T, et al. Human cardiac stem cells. ProcNatl Acad Sci U S A 2007;104:14068–73.

9. Schelbert HR, Inubushi M, Ross RS. PET imaging in small animals.J Nucl Cardiol 2003;10:513–20.

0. Lyngbaek S, Schneider M, Hansen JL, Sheikh SP. Cardiac regener-ation by resident stem and progenitor cells in the adult heart. Basic ResCardiol 2007;102:101–14.

1. Oyama T, Nagai T, Wada H, et al. Cardiac side population cells havea potential to migrate and differentiate into cardiomyocytes in vitroand in vivo. J Cell Biol 2007;176:329–41.

2. Rosenblatt-Velin N, Lepore MG, Cartoni C, Beermann F, PedrazziniT. FGF-2 controls the differentiation of resident cardiac precursorsinto functional cardiomyocytes. J Clin Invest 2005;115:1724–33.

3. Tateishi K, Ashihara E, Takehara N, et al. Clonally amplified cardiacstem cells are regulated by Sca-1 signaling for efficient cardiovascular

regeneration. J Cell Sci 2007;120:1791–800.
Page 12: Imaging Survival and Function of Transplanted Cardiac ... · BM-MSC bone marrow-derived mesenchymal stem cell CSC cardiac stem cell(s) DAPI 4=6-diamidino-2-phenylindole DiI-ac-LDL

3

3

3

3

3

3

4

4

4

4

4

4

Kd

F

1240 Li et al. JACC Vol. 53, No. 14, 2009Transplantation of Cardiac Resident Stem Cells April 7, 2009:1229–40

4. Garry DJ, Martin CM. Cardiac regeneration: self-service at the pump.Circ Res 2004;95:852–4.

5. McLaughlin L, Zhu G, Mistry M, et al. Apolipoprotein J/clusterinlimits the severity of murine autoimmune myocarditis. J Clin Invest2000;106:1105–13.

6. Zhang M, Methot D, Poppa V, Fujio Y, Walsh K, Murry CE.Cardiomyocyte grafting for cardiac repair: graft cell death and anti-death strategies. J Mol Cell Cardiol 2001;33:907–21.

7. van der Bogt KE, Schrepfer S, Sheikh AY, et al. Comparison oftransplantation of adipose tissue- and bone marrow-derived mesenchymalstem cells in the infarcted heart. Transplantation 2009;87:642–52.

8. Cao F, Wagner RA, Wilson KD, et al. Transcriptional and functionalprofiling of human embryonic stem cell-derived cardiomyocytes. PLoSONE 2008;3:e3474.

9. Mendell JR, Kissel JT, Amato AA, et al. Myoblast transfer in thetreatment of Duchenne’s muscular dystrophy. N Engl J Med 1995;333:832–8.

0. Wollert KC, Drexler H. Clinical applications of stem cells for theheart. Circ Res 2005;96:151–63.

1. Gnecchi M, He H, Liang OD, et al. Paracrine action accounts formarked protection of ischemic heart by Akt-modified mesenchymal

2. Cao F, Sadrzadeh Rafie AH, Abilez OJ, et al. In vivo imaging andevaluation of different biomatrices for improvement of stem cellsurvival. J Tissue Eng Regen Med 2007;1:465–8.

3. Laflamme MA, Chen KY, Naumova AV, et al. Cardiomyocytes derivedfrom human embryonic stem cells in pro-survival factors enhance functionof infarcted rat hearts. Nat Biotechnol 2007;25:1015–24.

4. Muraski JA, Rota M, Misao Y, et al. Pim-1 regulates cardiomyocytesurvival downstream of Akt. Nat Med 2007;13:1467–75.

5. Tillmanns J, Rota M, Hosoda T, et al. Formation of large coronaryarteries by cardiac progenitor cells. Proc Natl Acad Sci U S A2008;105:1668–73.

ey Words: molecular imaging y cardiac stem cells y ischemic heartisease y differentiation y cell fate.

APPENDIX

or the Supplemental Methods section, figures,

stem cells. Nat Med 2005;11:367–8. tables, and videos, please see the online version of this article.