nih public access domenico d’amario joão ferreira … circ res. 2011.pdf · qrt-pcr and western...

23
The Ephrin A1-EphA2 System Promotes Cardiac Stem Cell Migration after Infarction Polina Goichberg 1 , Yingnan Bai 1 , Domenico D’Amario 1 , João Ferreira-Martins 1 , Claudia Fiorini 2 , Hanqiao Zheng 1 , Sergio Signore 1 , Federica del Monte 3 , Sergio Ottolenghi 4 , David D’Alessandro 2 , Robert E. Michler 2 , Toru Hosoda 1 , Piero Anversa 1 , Jan Kajstura 1 , Marcello Rota 1 , and Annarosa Leri 1 1 Departments of Anesthesia and Medicine, and Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115 2 Montefiore Medical Center, Albert Einstein College of Medicine, New York, NY 3 Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115 4 Department of Biotechnology and Bioscience, University of Milano-Bicocca, Milan, Italy Abstract Rationale—Understanding the mechanisms that regulate trafficking of human cardiac stem cells (hCSCs) may lead to development of new therapeutic approaches for the failing heart. Objectives—We tested whether the motility of hCSCs in immunosuppressed infarcted animals is controlled by the guidance system that involves the interaction of Eph receptors with ephrin ligands. Methods and Results—Within the cardiac niches, cardiomyocytes expressed preferentially the ephrin A1 ligand, while hCSCs possessed the EphA2 receptor. Treatment of hCSCs with ephrin A1 resulted in the rapid internalization of the ephrin A1-EphA2 complex, post-translational modifications of Src kinases, and morphological changes consistent with the acquisition of a motile cell phenotype. Ephrin A1 enhanced the motility of hCSCs in vitro, and their migration in vivo following acute myocardial infarction. At two weeks after infarction, the volume of the regenerated myocardium was two-fold larger in animals injected with ephrin A1-activated hCSCs than in animals receiving control hCSCs; this difference was dictated by a greater number of newly formed cardiomyocytes and coronary vessels. The increased recovery in myocardial mass with ephrin A1-treated hCSCs was characterized by further restoration of cardiac function and by a reduction in arrhythmic events. Conclusions—Ephrin A1 promotes the motility of EphA2-positive hCSCs, facilitates their migration to the area of damage, and enhances cardiac repair. Thus, in situ stimulation of resident hCSCs with ephrin A1 or their ex vivo activation prior to myocardial delivery improves cell Address for Correspondence: Annarosa Leri, M.D., Departments of Anesthesia and Medicine, and Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115; phone: 617-5258174; [email protected]. Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. Disclosures None. NIH Public Access Author Manuscript Circ Res. Author manuscript; available in PMC 2012 April 29. Published in final edited form as: Circ Res. 2011 April 29; 108(9): 1071–1083. doi:10.1161/CIRCRESAHA.110.239459. NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript

Upload: hakhue

Post on 19-Jul-2018

220 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: NIH Public Access Domenico D’Amario João Ferreira … Circ Res. 2011.pdf · qRT-PCR and Western Blotting ... mice were injected with hCSCs pre-treated with ephrin A1 or Fc for

The Ephrin A1-EphA2 System Promotes Cardiac Stem CellMigration after Infarction

Polina Goichberg1, Yingnan Bai1, Domenico D’Amario1, João Ferreira-Martins1, ClaudiaFiorini2, Hanqiao Zheng1, Sergio Signore1, Federica del Monte3, Sergio Ottolenghi4, DavidD’Alessandro2, Robert E. Michler2, Toru Hosoda1, Piero Anversa1, Jan Kajstura1, MarcelloRota1, and Annarosa Leri11Departments of Anesthesia and Medicine, and Division of Cardiovascular Medicine, Brighamand Women’s Hospital, Harvard Medical School, Boston, MA 021152Montefiore Medical Center, Albert Einstein College of Medicine, New York, NY3Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School,Boston, MA 021154Department of Biotechnology and Bioscience, University of Milano-Bicocca, Milan, Italy

AbstractRationale—Understanding the mechanisms that regulate trafficking of human cardiac stem cells(hCSCs) may lead to development of new therapeutic approaches for the failing heart.

Objectives—We tested whether the motility of hCSCs in immunosuppressed infarcted animals iscontrolled by the guidance system that involves the interaction of Eph receptors with ephrinligands.

Methods and Results—Within the cardiac niches, cardiomyocytes expressed preferentially theephrin A1 ligand, while hCSCs possessed the EphA2 receptor. Treatment of hCSCs with ephrinA1 resulted in the rapid internalization of the ephrin A1-EphA2 complex, post-translationalmodifications of Src kinases, and morphological changes consistent with the acquisition of amotile cell phenotype. Ephrin A1 enhanced the motility of hCSCs in vitro, and their migration invivo following acute myocardial infarction. At two weeks after infarction, the volume of theregenerated myocardium was two-fold larger in animals injected with ephrin A1-activated hCSCsthan in animals receiving control hCSCs; this difference was dictated by a greater number ofnewly formed cardiomyocytes and coronary vessels. The increased recovery in myocardial masswith ephrin A1-treated hCSCs was characterized by further restoration of cardiac function and bya reduction in arrhythmic events.

Conclusions—Ephrin A1 promotes the motility of EphA2-positive hCSCs, facilitates theirmigration to the area of damage, and enhances cardiac repair. Thus, in situ stimulation of residenthCSCs with ephrin A1 or their ex vivo activation prior to myocardial delivery improves cell

Address for Correspondence: Annarosa Leri, M.D., Departments of Anesthesia and Medicine, and Division of CardiovascularMedicine, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115; phone: 617-5258174;[email protected]'s Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to ourcustomers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review ofthe resulting proof before it is published in its final citable form. Please note that during the production process errors may bediscovered which could affect the content, and all legal disclaimers that apply to the journal pertain.DisclosuresNone.

NIH Public AccessAuthor ManuscriptCirc Res. Author manuscript; available in PMC 2012 April 29.

Published in final edited form as:Circ Res. 2011 April 29; 108(9): 1071–1083. doi:10.1161/CIRCRESAHA.110.239459.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 2: NIH Public Access Domenico D’Amario João Ferreira … Circ Res. 2011.pdf · qRT-PCR and Western Blotting ... mice were injected with hCSCs pre-treated with ephrin A1 or Fc for

targeting to sites of injury, possibly providing a novel strategy for the management of the diseasedheart.

Keywordsephrin; cardiac stem cells; myocardial regeneration; cell migration

The identification of cardiac stem cells (CSCs) and their niches in the mammalian heart1–4

has changed our understanding of the mechanisms of myocardial homeostasis and tissuerepair following injury. The adult heart is now considered a self-renewing organ regulatedby a resident stem cell compartment, which controls myocyte turnover physiologically andcontributes to myocardial recovery in the presence of damage.5,6 This revolutionary view ofmyocardial biology has raised the important question concerning the processes thatmodulate CSC migration and differentiation. The initial events, involving CSC activation,commitment and acquisition of a motile cell phenotype, are triggered by myocytes andfibroblasts within the niches, where they function as supporting cells.7 The cross-talkbetween adjacent cells in the niche microenvironment conditions the fate of CSCs,according to the need of the organ and the preservation of its structural and functionalintegrity.8–10

Interaction of Eph receptor tyrosine kinases with membrane-bound ephrin ligands is part ofa complex contact-dependent communication between neighboring cells.11,12 The ephrin-Eph complex on the plasma membrane of adjacent cells results in bi-directional signaling,ultimately, modulating cell adhesion or movement, division or differentiation.12 The ephrinligand family is divided in two classes, A (A1–5) and B (B1–3), based on their structure andability to bind to the nine EphA and five EphB receptors, respectively.12 The ephrin-Ephaxis regulates self-renewal or commitment, adhesion or migration of stem cells in variousorgans, including the brain, the bone marrow and the intestine.13,14 Importantly, Ephreceptor tyrosine kinases and their ligands participate in the formation of the outflow tractand coronary vessels during cardiac development.15

These observations raise the possibility that the ephrin-Eph system may be operative inCSCs where it may have a positive or negative effect on the cardiac repair process.Currently, phase 1 clinical trials are in progress and human CSCs (hCSCs) are delivered topatients with acute and chronic ischemic cardiomyopathy (ClinicalTrials.gov Identifiers:NCT00474461; NCT00893360), emphasizing the relevance that pathways favoring homingand translocation of the delivered cells may have on patients’ outcome. Therefore, the roleof the ephrin-Eph complex in the fate of mouse and human CSCs was determined to definethe potential therapeutic impact of this system on myocardial regeneration after infarction.

MethodsAn expanded Methods section is available in the Online Data Supplement.

Human Cardiac Stem Cells (hCSCs) and CardiomyocytesDiscarded myocardial samples from patients who underwent elective cardiac surgery wereenzymatically dissociated and c-kit-positive hCSCs were obtained;2 expanded hCSCs wereemployed for in vitro and in vivo studies. Explanted human hearts and donor hearts declinedfor transplantation were utilized to obtain cardiomyocytes.

Mouse CSCs (mCSCs) and CardiomyocytesThe heart of C57BL/6 mice at 3 months of age was perfused with a collagenase solution andmyocytes and c-kit-positive mCSCs were isolated.7,8

Goichberg et al. Page 2

Circ Res. Author manuscript; available in PMC 2012 April 29.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 3: NIH Public Access Domenico D’Amario João Ferreira … Circ Res. 2011.pdf · qRT-PCR and Western Blotting ... mice were injected with hCSCs pre-treated with ephrin A1 or Fc for

qRT-PCR and Western BlottingRNA was extracted from hCSCs, mCSCs, and human and mouse myocytes for themeasurement of ephrin ligand and Eph receptor transcripts.8,10 Protein lysates of hCSCs,human myocytes, and mouse heart tissue were utilized for the assessment of EphA2, ephrinA1 and phosphorylated Src kinases.8–10 For primers and antibodies, see Online Tables I andII, respectively.

ImmunocytochemistryFollowing fixation in 4% paraformaldehyde, hCSCs and human cardiomyocytes wereexposed to antibodies2 recognizing c-kit, EphA2, ephrin A1 and sarcomeric proteins. Cellmorphology was defined by incubation with phalloidin. Ephrin A1-hFcγ binding wasidentified with TRITC-conjugated anti human Fcγ antibody.

Adhesion and Migration AssayCalcein-labeled hCSCs were plated on ephrin A1-coated wells and the fraction of adheredhCSCs was estimated by measuring fluorescence intensity. hCSCs, exposed to ephrin A1-hFcγ (ephrin A1) or human IgG (Fc), were allowed to migrate towards Hepatocyte growthfactor (HGF) in a transwell system.9 The number of transmigrated cells was counted by flowcytometry.

Myocardial InfarctionMyocardial infarction was produced in c-kit-EGFP mice; ephrin A1 was administered to theborder zone, and the number of c-kit-EGFP-positive CSCs measured.2,8 Infarcted C57BL/6mice were injected with hCSCs pre-treated with ephrin A1 or Fc for the ex vivo analysis ofhCSC motility by two-photon microscopy.9 Immunosuppressed infarcted Fischer 344 ratswere sacrificed two weeks after surgery and delivery of hCSCs for the evaluation ofmyocardial regeneration. Sham-operated and infarcted rats injected with PBS were used ascontrols. At sacrifice ventricular function was determined. The heart was then fixed forhistological analysis. 2,8 Newly formed cardiomyocytes and coronary vessels wererecognized by EGFP labeling and in situ hybridization for Alu probe and Y-chromosome.2Specificity of staining was determined by spectral analysis.6 A protocol of programmedelectrical stimulation was used to assess the propensity of the infarcted rat heart to developventricular arrhythmias.

Statistical AnalysisResults are presented as mean±SD. Significance between two comparisons was determinedby unpaired Student’s t-test and among multiple comparisons by the Bonferronimethod.2,7–9 All P values are two-sided and values less than 0.05 were consideredsignificant. See Online Table III for sampling.

ResultsEphA2 and Ephrin A1 in the Myocardium

C-kit positive hCSCs are organized in niches which are located preferentially in the atria andapex.2,16 hCSCs are functionally connected to cardiomyocytes, which act as supporting cellsand influence the fate of adjacent primitive cells. The components of this cell-to-cellinteraction within the cardiac niches are largely unknown.8 In analogy to other self-renewingorgans,13,14,17 the Eph-ephrin system may regulate the motility of resident stem cells withinthe heart. Therefore, the presence of the Eph-ephrin family members was measured by qRT-PCR in hCSCs and human cardiomyocytes to search for gene products differentiallyexpressed in these two cell classes (Online Figure I). EphA2 receptor mRNA was abundant

Goichberg et al. Page 3

Circ Res. Author manuscript; available in PMC 2012 April 29.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 4: NIH Public Access Domenico D’Amario João Ferreira … Circ Res. 2011.pdf · qRT-PCR and Western Blotting ... mice were injected with hCSCs pre-treated with ephrin A1 or Fc for

in hCSCs, while transcripts of the ephrin A1 ligand were highly represented incardiomyocytes (Figure 1A). A similar distribution was found in mCSCs and mousecardiomyocytes. The two distinct isoforms of the ligand, ephrin A1a and ephrin A1b, wereequally represented in the myocardium (Online Figure I), but the effect of these two proteinson EphA2 receptor activation is, at present, unknown. The preferential localization ofEphA2 in hCSCs and ephrin A1 in human cardiomyocytes was confirmed by Westernblotting (Figure 1B), and immunolabeling and confocal microscopy (Figure 1C and 1D).

To establish whether these findings in isolated hCSCs and cardiomyocytes mimicked thetissue properties, the expression of EphA2 and ephrin A1 was determined in the humanmyocardium. Clusters of hCSCs were nested in the interstitium and were coupled withneighboring myocytes by the expression of connexin 43 and N-cadherin (Figure 1E). Theephrin A1 ligand was present in myocytes adjacent to EphA2-positive hCSCs (Figure 1F).Importantly, ephrin A1 was restricted to the myocyte compartment; it was not detected inendothelial cells (ECs), smooth muscle cells (SMCs) or fibroblasts (Figure 1G). These dataraise the possibility that cardiomyocytes carrying the ephrin A1 ligand interact with hCSCspossessing the EphA2 receptor and, as a result, modify their motile phenotype within thecardiac niches.

Ephrin A1 and hCSC MotilityCardiomyocytes may influence the behavior of hCSCs by direct cell-to-cell contact or bysecretion of a soluble signal. Thus, the functional role of the ephrin A1-EphA2 axis wasestablished in vitro by exposing EphA2-positive hCSCs to a human ephrin A1-Fcγ chimericprotein (ephrin A1), or control human IgG (Fc). The rapid adhesion of hCSCs to ephrin A1-coated surfaces documented the functional competence of the EphA2 receptor. Transfectionwith siRNA specific for EphA2 decreased significantly its expression and was notaccompanied by a compensatory increase in the quantity of EphA3 and EphA4 transcripts(Figure 2A). EphA2 down-regulation abrogated the adhesive response of hCSCs toimmobilized ephrin A1 (Figure 2B). Ephrin A1 promoted rearrangement of the actincytoskeleton in hCSCs, changing their shape from a sessile to a motile state (Figure 2C).This change in cell morphology was characterized by a rapid internalization of the ephrinA1-EphA2 complex, from the plasma membrane to the cytoplasm (Figure 2D).

Since Src kinase represents a well-established downstream effector of Eph receptorsignaling,12 the state of phosphorylation of Src was evaluated in ephrin A1-stimulatedhCSCs. Following ligand binding, there was a time-dependent increase in thephosphorylation of the activatory site of Src family kinases at tyrosine 416, which wasassociated with a concomitant decrease in phosphorylation of the inhibitory site of Src attyrosine 527 (Figure 2E). These post-translational modifications of Src kinases indicate thatthe EphA2 pathway was activated in hCSCs exposed to ephrin A1.

The chemoattractant HGF favors the translocation of stem cells to sites of ischemicmyocardial damage.9,18 In the presence of HGF, hCSCs acquired a motile phenotypecharacterized by accumulation of EphA2 at the leading edge of migrating cells (Figure 2F).As expected, hCSCs moved towards HGF in a Transwell migration assay (Figure 2G and2H). Pre-stimulation of hCSCs with recombinant ephrin A1 enhanced the spontaneousmotility and chemotactic response of these cells to low concentrations of HGF. However,the additive effect of ephrin A1 on cell locomotion was no longer apparent when hCSCswere exposed to high quantities of HGF, which saturated the migratory machinery of thecells. Migration of hCSCs towards high concentrations of HGF was significantly decreasedwhen the expression of EphA2 was inhibited by siRNA. A similar effect was observed whenhCSCs were exposed to a chemical inhibitor of Src kinase activity (Figure 2G and 2H).Importantly, ephrin A1 did not alter the rate of proliferation and apoptosis of hCSCs (Online

Goichberg et al. Page 4

Circ Res. Author manuscript; available in PMC 2012 April 29.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 5: NIH Public Access Domenico D’Amario João Ferreira … Circ Res. 2011.pdf · qRT-PCR and Western Blotting ... mice were injected with hCSCs pre-treated with ephrin A1 or Fc for

Figure II). Additionally, treatment with ephrin A1 did not modify the ability of hCSCs tocommit to cardiovascular lineages as shown by qRT-PCR and immunolabeling. The fractionof hCSCs expressing, α-sarcomeric actin (α-SA), α-smooth muscle actin (α-SMA) and vonWillebrand factor (vWF) was comparable in Fc-exposed and ephrin A1-treated cells (Figure2I). Collectively, these observations emphasize the critical role that the ephrin A1-EphA2system has in the migratory ability of hCSCs.

Endogenous Stem Cells and Myocardial InfarctionThe in vitro results raised the possibility that the ephrin A1-EphA2 axis enhances themigration of endogenous stem cells to the injured myocardium, favoring the recovery of theinfarcted heart. Two days after coronary artery occlusion in the mouse, the expression ofephrin A1 markedly increased in the border zone and distant myocardium (Figure 3A and3B). However, phosphorylation of the activatory site of Src family kinases at tyrosine 416did not differ in these two regions (Figure 3B). The myocardium is composed predominantlyof cardiomyocytes, which do not express the EphA2 receptor, preventing the activation ofdistal pathways. Ephrin A1 was found to be upregulated in human myocytes of explantedfailing hearts (Figure 3C), suggesting that the synthesis of ephrin A1 by the musclecompartment constitutes a relevant adaptive response aiming at the recruitment of hCSCs atsites of tissue damage. However, this regenerative process is restricted to the survivingregion of the ventricular wall,19 contributing minimally to the reconstitution of the infarctedmyocardium.8

To determine whether ephrin A1 positively affects the motility of EphA2-positive CSCs invivo, a transgenic mouse model in which the expression of enhanced green fluorescentprotein (EGFP) is driven by the c-kit promoter was employed.20 Ephrin A1 wasadministered in the border zone of acutely infarcted mice and the number of CSCs present inproximity of the necrotic tissue was measured 2 days later. Infarcted mice injected with Fcwere used as controls. In comparison with Fc-treated mice, the intramyocardial delivery ofephrin A1 resulted in a 2-fold increase in the number of c-kit-EGFP-positive CSCs (Figure4A and 4B).

Several factors had to be considered in the interpretation of these results. The accumulationof stem cells in the presence of ephrin A1 may involve enhanced recruitment, increased stemcell division, reduced apoptosis, or a combination of these variables. However, the fractionof cycling Ki67-positive CSCs was found to be comparable in ephrin A1- and Fc- treatedinfarcted hearts. Similarly, apoptosis of CSCs, measured by the TdT assay, did not differwith ephrin A1 or its absence (Figure 4C). Thus, ephrin A1 favors the translocation andhoming of CSCs to the site of myocardial injury, enhancing the cellular processesresponsible for cardiac repair.

Ephrin A1 and Migration of hCSCs In VivoThe observations in the mouse heart pointed to a potential role of ephrin A1 in the activationof EphA2 in hCSCs, favoring their translocation to the damaged myocardium and theinitiation of a regenerative response. For this purpose, EGFP-positive hCSCs were exposedto ephrin A1 in vitro and, following delivery to the border zone of acutely infarcted mice,the movement of these hCSCs from the site of injection to the infarcted myocardium, wasmonitored by two-photon microscopy.

With respect to control cells, ephrin A1-activated hCSCs showed a 2.5-fold increase in thespeed of locomotion within the myocardium (Figure 5; Online Movie I). A greaterdisplacement in the trajectory of migrating hCSCs within the tissue was also found (Figure5B). The enhanced movement of hCSCs with ephrin A1 was associated with a 25% increase

Goichberg et al. Page 5

Circ Res. Author manuscript; available in PMC 2012 April 29.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 6: NIH Public Access Domenico D’Amario João Ferreira … Circ Res. 2011.pdf · qRT-PCR and Western Blotting ... mice were injected with hCSCs pre-treated with ephrin A1 or Fc for

in the number of migrating cells. Thus, the ephrin A1-EphA2 pathway positively influencesthe timing and degree of hCSC trafficking and the onset and, potentially, the extent of tissuerepair.

Ephrin A1 and Myocardial Regeneration by hCSCsTo establish the effects of ephrin A1-activated hCSCs on tissue regeneration, hCSCs wereexposed to the ephrin A1 ligand, prior to delivery to the region bordering the acutelyinfarcted myocardium in immunosuppressed rats. Infarcted hearts injected with PBS or withhCSCs incubated with Fc were used as controls. hCSCs were infected with a lentiviruscarrying EGFP for the in vivo tracking of the formed progeny. All animals were sacrificed 2weeks after coronary artery ligation and cell or PBS administration.

The infarcted myocardium occupied a large portion of the left ventricle (LV), extendingfrom the endocardial to the epicardial aspect of the wall. A thin layer of spared myocyteswas detected in close proximity to the endocardium. Large bundles of collagen and vascularprofiles replaced the necrotic tissue at 2 weeks (Figure 6A). Therapy with both activated andnon-activated hCSCs resulted in myocardial regeneration which interfered in part with thenegative consequences of the healing process and scar formation (Figure 6B and 6C).Cardiac repair consisted of clusters of closely packed human cardiomyocytes and coronaryvessels (Figure 6D–6F). The level of myocyte formation, measured by the cell cycle proteinKi67, was comparable in these two groups of hearts, documenting that myocyte regenerationwas ongoing at sacrifice (Online Figure III). The human origin of the regenerated structureswas determined by the expression of EGFP, human DNA sequences with an Alu probe, anddetection of human Y-chromosome (Figure 6E and 6F). The specificity of immunolabelingfor EGFP and Alu was confirmed by spectral analysis (Figure 6G and 6H).

To evaluate quantitatively the efficacy of ephrin A1 activation of hCSCs, infarct size, thenumber of newly-formed myocytes, and their volume were determined and interpreted inrelation to ventricular hemodynamics. These parameters were then compared with thecorresponding values in infarcted hearts injected with hCSCs exposed to Fc. Coronaryligation resulted in an average 46% loss of LV myocytes in infarcted hearts injected withPBS, Fc-hCSCs and ephrin A1-hCSCs (Figure 7A).

Myocardial regeneration was detected in the two cell-treated groups. However, theaggregate volume of human myocytes was 2-fold larger in animals injected with ephrin A1-activated hCSCs than in rats receiving Fc-hCSCs. The volume of newly formed myocytes inthe two groups was comparable, indicating that the increase in myocyte number wasresponsible for the higher degree of myocardial regeneration with ephrin A1-treated hCSCs(Figure 7B). The increase in the length density of resistance arterioles and capillariesfollowed a similar pattern (Figure 7C and 7D), supporting the notion that hCSCs stimulatedby ephrin A1 differentiated in all cardiac cell lineages, retaining their multipotent phenotypein vivo.

Following treatment with ephrin A1-hCSCs, cardiac repair resulted in a 37% reduction ofinfarct size. This value was nearly 2-fold larger than that measured after the injection of Fc-hCSCs (Figure 7E). The recovery in myocardial mass mediated by ephrin A1-hCSCs wascoupled with a less pronounced increase of LV end-diastolic pressure (LVEDP), anddecrease in +dP/dt and −dP/dt after infarction; however, these changes did not reachstatistical significance. LV developed pressure (LVDevP) and ejection fraction (EF) werehigher in infarcted hearts treated with ephrin A1-activated hCSCs and these differences weresignificant (Figure 8A).

Goichberg et al. Page 6

Circ Res. Author manuscript; available in PMC 2012 April 29.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 7: NIH Public Access Domenico D’Amario João Ferreira … Circ Res. 2011.pdf · qRT-PCR and Western Blotting ... mice were injected with hCSCs pre-treated with ephrin A1 or Fc for

Cardiac Repair and Ventricular ArrhythmiaA critical issue in need of resolution concerned whether the inhomogeneity of themyocardium after infarction was enhanced further by regeneration and the formation ofsmall neonatal-like cardiomyocytes, potentiating the incidence of arrhythmia. Thus,infarcted hearts, injected with PBS, Fc-hCSCs and ephrin A1-hCSCs, were studied ex vivoin a Langendorff preparation. To mimic the in vivo results, this analysis was conducted twoweeks after coronary artery ligation and cells or PBS delivery.

Perfused hearts were subjected to programmed electrical stimulation to induce ventriculartachycardia and fibrillation.21 Arrhythmic events were not detected in sham-operated hearts;88% of infarcted non-treated hearts showed episodes of arrhythmia. This value decreased to45% following treatment with Fc-hCSCs. In comparison with infarcted hearts injected withPBS and Fc-hCSCs, therapy by ephrin A1-hCSCs decreased the frequency of arrhythmia by89% and 78%, respectively (Figure 8B). Since the size of myocytes was comparable in cell-treated hearts, the greater degree of myocardial regeneration and infarct size reduction withephrin A1-hCSCs may account for the difference in arrhythmia between the two groups ofcell-treated infarcted hearts.

DiscussionThe results of the present study indicate that the ephrin A1-EphA2 system in mouse andhuman CSCs promotes a selective response, which involves cell migration, without affectingcell division, survival and differentiation. This specific function of the ephrin A1-EphA2axis in CSCs was documented by a series of in vitro and in vivo assays, which demonstrateda comparable motile effect on the endogenous and delivered CSCs. This critical role of theephrin A1-EphA2 complex in the migratory properties of mouse and human CSCs wasidentified by the analysis of the differential expression of ephrin ligands and Eph receptorsin the myocyte compartment and primitive cell pool, respectively. Based on this premise, wefound that ephrin A1 in cardiomyocytes and EphA2 in CSCs are implicated in cell-to-cellcommunication between undifferentiated and supporting cells within the niches, enhancingCSC migration. Importantly, ephrin A1 activation of hCSCs prior to their injection inproximity of the infarcted myocardium of immunosuppressed animals resulted in a two-foldincrease in myocardial regeneration and improved the recovery of ventricular function.

In this regard, the relevance of the ephrin/Eph system in the adult heart has never beenshown. Similarly, the existence and function of this axis in mouse and human CSCs andmyocardial regeneration was previously unknown. Migration of primitive cells representsthe crucial prerequisite for efficient repair of damaged myocardium, emphasizing thesignificance of novel strategies aiming at the enhancement of CSC recruitment at the site ofischemic injury. The dichotomic pattern of ephrin A1 and EphA2 protein distributionrepresents a classical model of ligand-receptor interaction, which enables thecommunication between CSCs and myocytes in the cardiac niches, impacting on myocardialregeneration.

EphA2, Ephrin A1 and Cardiac RepairThe injection of hCSCs after infarction resulted over a period of 14 days in a significantreduction of infarct size mediated by their differentiation into a large number ofcardiomyocytes, resistance coronary arterioles and capillary structures. Although markedlyincreased by ephrin A1 activation of hCSCs, the process of myocardial regeneration retainedthe same proportion between newly-formed cardiomyocytes and coronary vasculature,indicating that the multipotentiality of the injected hCSCs was not affected by the ephrin A1ligand. The similarity in volume composition of the new myocardium in the two groups of

Goichberg et al. Page 7

Circ Res. Author manuscript; available in PMC 2012 April 29.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 8: NIH Public Access Domenico D’Amario João Ferreira … Circ Res. 2011.pdf · qRT-PCR and Western Blotting ... mice were injected with hCSCs pre-treated with ephrin A1 or Fc for

cell-treated infarcted hearts supports the notion that hCSCs recruited at the site of injuryunderwent comparable clonal activation22 giving rise to an equivalent number ofcardiomyocytes and vascular cells. The larger magnitude of myocardium generated byephrin A1-treated hCSCs emphasizes the relevance of cell migration for the efficient repairof the damaged heart. Importantly, downregulation of EphA2 in hCSCs and blockade of thereceptor downstream signaling impairs the motility of hCSCs.

A tight balance between myocyte size and capillary and arteriolar density23 has to bemaintained to ensure an adequate blood flow and oxygenation of the regeneratedmyocardium. The principal structural variables of the microvasculature of the heart that arefunctionally relevant to the oxygenation of the newly formed tissue are capillary luminalvolume density, capillary luminal surface density, and the average diffusion distance fromthe capillary wall to the surrounding tissue.24 These parameters are strictly dependent on thenumber of capillaries per unit area of myocardium; in the regenerated tissue, there was onecapillary every ~20 cardiomyocytes, mimicking the pattern of tissue oxygenation presentearly after birth in rodents.24 Similarly, the size of cardiomyocytes was comparable to thatfound at birth in the mouse and rat heart.24 The capillary-to-myocyte ratio increases withpostnatal maturation, reaching a value of nearly one in the young adult heart. Whether theregenerated myocytes and vascular structures will reach with time the adult cell phenotype isopen to question. It is also currently unknown whether ephrin A1 favorably affects thisprocess of maturation.

Ephrin A1-EphA2 System and Myocardial RegenerationThe results of the present study indicate that myocardial infarction results in a significantupregulation of ephrin A1 in the surviving myocytes and this response is greater in theborder zone than in the remote myocardial region. The expression of IGF-1 and HGFtranscripts increases in the spared tissue acutely after infarction.25,26 However, theconcentration of HGF and IGF-1 proteins remains relatively constant in the border zone,remote myocardium and ischemic region at 6–12 hours after coronary artery ligation,suggesting that these two growth factors undergo rapid turnover.27 Although the synthesis ofephrin A1 is enhanced after infarction, the administration of the exogenous ligand isnecessary to induce activation and efficient recruitment of CSCs. IGF-1 and HGF, togetherwith ephrin A1, bind to the corresponding receptors on resident CSCs, promoting division,survival, and translocation of these cells towards the necrotic tissue.9,18 This possibility issupported by the increase in number of CSCs in proximity of the infarct shortly aftercoronary occlusion in animals and humans.8 Ephrin A1 and HGF are both highly effective inpromoting cell movement. However, the positive effect of HGF on CSC migration requiresthe activation of the cell motile state by ephrin A1, which enhances the susceptibility ofCSCs to translocate to the site of injury. The motility of hCSCs within the border zone of theinfarcted heart was two-fold higher in animals treated with ephrin A1, suggesting that in situactivation of resident hCSCs with this ligand, or their ex vivo manipulation, may improvecell targeting to sites of damage, providing a novel strategy for the management of thepathologic heart.

The augmented expression of ephrin A1 in mouse myocytes after infarction and in humanmyocytes from patients with end-stage heart failure indicate that, under conditions of stress,the heart initiates a series of compensatory processes aiming at the mobilization of CSCstowards the area of damage. In analogy with the myocardium, hypoxia and inflammationenhance the expression of ephrin A1 and EphA2 in the skin and vascular structures.28 Animportant question left unanswered by this and previous studies concerns the inability of thepool of resident CSCs to spontaneously induce a regenerative response and reestablish thestructural and functional integrity of the infarcted heart. This mechanism is very effective inlower vertebrates29 but it is not efficient in the adult heart of mammals, including humans.19

Goichberg et al. Page 8

Circ Res. Author manuscript; available in PMC 2012 April 29.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 9: NIH Public Access Domenico D’Amario João Ferreira … Circ Res. 2011.pdf · qRT-PCR and Western Blotting ... mice were injected with hCSCs pre-treated with ephrin A1 or Fc for

CSCs within the infarct die by apoptosis and the formation of myocytes and coronaryvessels is restricted to the unaffected portion of the ventricular wall, with minimal or noneregeneration of the infarcted tissue. Whether the CSCs nested in the spared myocardium arenot equipped to sense signals from the infarct, or growth stimuli from the infarct areinadequate to trigger the migration of these primitive cells, or both, is presently unknown.

EphA2 and Ephrin A1 in CSCs and CardiomyocytesConsistent with the reciprocal distribution of EphA2 receptor and ephrin A1 ligand in CSCsand cardiomyocytes, progenitor cells in the hippocampus13 and intestinal crypt14 areenriched in Eph receptor, while the surrounding differentiated cells exhibit high levels ofephrin ligands. The distinct expression pattern of EphA2 and ephrin A1 in CSCs andcardiomyocytes may generate a bidirectional signaling that provide positional guidance cuesto maintain tissue homeostasis and favor cardiac repair. In all tissues, the ephrin-Eph systemuses common effectors including the Src family of kinases,12 which, as shown here inhCSCs following ephrin A1 stimulation, experience rapid post-translational modifications.

The transient net increase in Src family kinase activity in hCSCs with ephrin A1 wascoupled with rearrangement of the actin cytoskeleton and cell shape polarization, whichresulted in enhanced spontaneous and HGF-induced motility of hCSCs. These molecularand morphological adaptations were dictated by the internalization of the ephrin A1-EphA2complex from the cell membrane to the cell cytoplasm. Ligand-induced Eph receptorendocytosis is a critical determinant of the transition from the high affinity cell-to-celladhesion to the repulsive response.12

In addition to cell migration, the ephrin-Eph family of proteins controls multiple cellularfunctions, including growth, differentiation, and death of stem/progenitor cells.11,12

Surprisingly, ephrin A1 did not affect the rate of proliferation, commitment and apoptosis ofhCSCs in vitro and in vivo, pointing to a restricted function of the ephrin A1-EphA2 axis inthe regulation of the movement of human and mouse CSCs. The consequences of Ephactivation on progenitor cell fate are ligand, cell context, and kinase activity dependent.17,30

In physiological conditions, the ephrin-Eph signaling ensures a proper balance between astem cell and its progeny and may interfere or favor cell survival, replication andcommitment. In the brain, distinct components of the ephrin-Eph family have oppositeeffects on apoptosis and proliferation of neural stem cells.13,31

Reliance or independence of Eph receptor signaling from the ligand, or downstream kinaseactivity, leads to a wide array of contrasting effects on stem cell behavior.17,30 The spatial-mechanical characteristics of the interaction between ephrin A1 and EphA2 moleculesinfluence receptor function and the cell migratory response.32 The geometry of inter-membrane signaling may play a major role in the heart in which electromechanicallyconnected myocytes form a functional syncytium. Cardiomyocytes are subjected toconsiderable physical forces which impact, in an unknown manner, on structurally joinedCSCs. The increased loading state on the surviving myocardium after infarction may berelevant in the upregulation of the molecular pathways responsible for ephrin A1 synthesisand activation of resident CSCs.

Supplementary MaterialRefer to Web version on PubMed Central for supplementary material.

Goichberg et al. Page 9

Circ Res. Author manuscript; available in PMC 2012 April 29.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 10: NIH Public Access Domenico D’Amario João Ferreira … Circ Res. 2011.pdf · qRT-PCR and Western Blotting ... mice were injected with hCSCs pre-treated with ephrin A1 or Fc for

Non-Standard Abbreviations and Acronyms

CSC cardiac stem cells

hCSCs Human CSCs

mCSCs Mouse CSCs

CM Cardiomyocytes

hCM Human CM

mCM Mouse CM

HGF hepatocyte growth factor

ECs endothelial cells

SMCs smooth muscle cells

α-SA α-sarcomeric actin

α-SMA α-smooth muscle actin

vWf von Willebrand factor

AcknowledgmentsSources of Funding

This work is supported by NIH.

References1. Beltrami AP, Barlucchi L, Torella D, Baker M, Limana F, Chimenti S, Kasahara H, Rota M, Musso

E, Urbanek K, Leri A, Kajstura J, Nadal-Ginard B, Anversa P. Adult cardiac stem cells aremultipotent and support myocardial regeneration. Cell. 2003; 114:763–776. [PubMed: 14505575]

2. Bearzi C, Rota M, Hosoda T, Tillmanns J, Nascimbene A, De Angelis A, Yasuzawa-Amano S,Trofimova I, Siggins RW, Lecapitaine N, Cascapera S, Beltrami AP, D'Alessandro DA, Zias E,Quaini F, Urbanek K, Michler RE, Bolli R, Kajstura J, Leri A, Anversa P. Human cardiac stemcells. Proc Natl Acad Sci USA. 2007; 104:14068–14073. [PubMed: 17709737]

3. Smith RR, Barile L, Cho HC, Leppo MK, Hare JM, Messina E, Giacomello A, Abraham MR,Marban E. Regenerative potential of cardiosphere-derived cells expanded from percutaneousendomyocardial biopsy specimens. Circulation. 2007; 115:896–908. [PubMed: 17283259]

4. Pfister O, Oikonomopoulos A, Sereti KI, Sohn RL, Cullen D, Fine GC, Mouquet F, Westerman K,Liao R. Role of the ATP-binding cassette transporter Abcg2 in the phenotype and function ofcardiac side population cells. Circ Res. 2008; 103:825–835. [PubMed: 18787193]

5. Bergmann O, Bhardwaj RD, Bernard S, Zdunek S, Barnabe-Heider F, Walsh S, Zupicich J, AlkassK, Buchholz BA, Druid H, Jovinge S, Frisen J. Evidence for cardiomyocyte renewal in humans.Science. 2009; 324:98–102. [PubMed: 19342590]

6. Kajstura J, Gurusamy N, Ogorek B, Goichberg P, Clavo-Rondon C, Hosoda T, D'Amario D,Bardelli S, Beltrami AP, Cesselli D, Bussani R, Del Monte F, Quaini F, Rota M, Beltrami CA,Buchholz BA, Leri A, Anversa P. Myocyte turnover in the aging human heart. Circ Res. 2010;107:1374–1386. [PubMed: 21088285]

7. Urbanek K, Cesselli D, Rota M, Nascimbene A, De Angelis A, Hosoda T, Bearzi C, Boni A, BolliR, Kajstura J, Anversa P, Leri A. Stem cell niches in the adult mouse heart. Proc Natl Acad SciUSA. 2006; 103:9226–9231. [PubMed: 16754876]

8. Boni A, Urbanek K, Nascimbene A, Hosoda T, Zheng H, Delucchi F, Amano K, Gonzalez A, VitaleS, Ojaimi C, Rizzi R, Bolli R, Yutzey KE, Rota M, Kajstura J, Anversa P, Leri A. Notch1 regulatesthe fate of cardiac progenitor cells. Proc Natl Acad Sci USA. 2008; 105:15529–15534. [PubMed:18832173]

Goichberg et al. Page 10

Circ Res. Author manuscript; available in PMC 2012 April 29.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 11: NIH Public Access Domenico D’Amario João Ferreira … Circ Res. 2011.pdf · qRT-PCR and Western Blotting ... mice were injected with hCSCs pre-treated with ephrin A1 or Fc for

9. Gonzalez A, Rota M, Nurzynska D, Misao Y, Tillmanns J, Ojaimi C, Padin-Iruegas ME, Muller P,Esposito G, Bearzi C, Vitale S, Dawn B, Sanganalmath SK, Baker M, Hintze TH, Bolli R, UrbanekK, Hosoda T, Anversa P, Kajstura J, Leri A. Activation of cardiac progenitor cells reverses thefailing heart senescent phenotype and prolongs lifespan. Circ Res. 2008; 102:597–606. [PubMed:18202313]

10. Urbanek K, Cabral-da-Silva MC, Ide-Iwata N, Maestroni S, Delucchi F, Zheng H, Ferreira-MartinsJ, Ogorek B, D'Amario D, Bauer M, Zerbini G, Rota M, Hosoda T, Liao R, Anversa P, Kajstura J,Leri A. Inhibition of Notch1-dependent cardiomyogenesis leads to a dilated myopathy in theneonatal heart. Circ Res. 2010; 107:429–441. [PubMed: 20558824]

11. Lackmann M, Boyd AW. Eph, a protein family coming of age: more confusion, insight, orcomplexity? Science Signal. 2008; 1:re2.

12. Pasquale EB. Eph-ephrin bidirectional signaling in physiology and disease. Cell. 2008; 133:38–52.[PubMed: 18394988]

13. Depaepe V, Suarez-Gonzalez N, Dufour A, Passante L, Gorski JA, Jones KR, Ledent C,Vanderhaeghen P. Ephrin signaling controls brain size by regulating apoptosis of neuralprogenitors. Nature. 2005; 435:1244–1250. [PubMed: 15902206]

14. Holmberg J, Genander M, Halford MM, Anneren C, Sondell M, Chumley MJ, Silvany RE,Henkemeyer M, Frisen J. EphB receptors coordinate migration and proliferation in the intestinalstem cell niche. Cell. 2006; 125:1151–1163. [PubMed: 16777604]

15. Kuijper S, Turner CJ, Adams RH. Regulation of angiogenesis by Eph-ephrin interactions. TrendsCardiovascul Med. 2007; 17:145–151.

16. Bearzi C, Leri A, Lo Monaco F, Rota M, Gonzalez A, Hosoda T, Pepe M, Qanud K, Ojaimi C,Bardelli S, D'Amario D, D'Alessandro DA, Michler RE, Dimmeler S, Zeiher AM, Urbanek K,Hintze TH, Kajstura J, Anversa P. Identification of a coronary vascular progenitor cell in thehuman heart. Proc Natl Acad Sci USA. 2009; 106:15885–15890. [PubMed: 19717420]

17. Genander M, Halford MM, Xu NJ, Eriksson M, Yu Z, Qiu Z, Martling A, Greicius G, Thakar S,Catchpole T, Chumley MJ, Zdunek S, Wang C, Holm T, Goff SP, Pettersson S, Pestell RG,Henkemeyer M, Frisen J. Dissociation of EphB2 signaling pathways mediating progenitor cellproliferation and tumor suppression. Cell. 2009; 139:679–692. [PubMed: 19914164]

18. Urbanek K, Rota M, Cascapera S, Bearzi C, Nascimbene A, De Angelis A, Hosoda T, Chimenti S,Baker M, Limana F, Nurzynska D, Torella D, Rotatori F, Rastaldo R, Musso E, Quaini F, Leri A,Kajstura J, Anversa P. Cardiac stem cells possess growth factor-receptor systems that afteractivation regenerate the infarcted myocardium, improving ventricular function and long-termsurvival. Circ Res. 2005; 97:663–673. [PubMed: 16141414]

19. Beltrami AP, Urbanek K, Kajstura J, Yan SM, Finato N, Bussani R, Nadal-Ginard B, Silvestri F,Leri A, Beltrami CA, Anversa P. Evidence that human cardiac myocytes divide after myocardialinfarction. N Engl J Med. 2001; 344:1750–1757. [PubMed: 11396441]

20. Cairns LA, Moroni E, Levantini E, Giorgetti A, Klinger FG, Ronzoni S, Tatangelo L, Tiveron C,De Felici M, Dolci S, Magli MC, Giglioni B, Ottolenghi S. Kit regulatory elements required forexpression in developing hematopoietic and germ cell lineages. Blood. 2003; 102:3954–3962.[PubMed: 12907433]

21. Roell W, Lewalter T, Sasse P, Tallini YN, Choi BR, Breitbach M, Doran R, Becher UM, HwangSM, Bostani T, von Maltzahn J, Hofmann A, Reining S, Eiberger B, Gabris B, Pfeifer A, Welz A,Willecke K, Salama G, Schrickel JW, Kotlikoff MI, Fleischmann BK. Engraftment of connexin43-expressing cells prevents post-infarct arrhythmia. Nature. 2007; 450:819–824. [PubMed:18064002]

22. Hosoda T, D'Amario D, Cabral-Da-Silva MC, Zheng H, Padin-Iruegas ME, Ogorek B, Ferreira-Martins J, Yasuzawa-Amano S, Amano K, Ide-Iwata N, Cheng W, Rota M, Urbanek K, KajsturaJ, Anversa P, Leri A. Clonality of mouse and human cardiomyogenesis in vivo. Proc Natl AcadSci USA. 2009; 106:17169–17174. [PubMed: 19805158]

23. Rakusan, K. Assessment of cardiac growth. In: Zak, R., editor. Growth of the heart in health anddisease. New York: Raven Press; 1984. p. 25-40.

24. Anversa, P.; Olivetti, G. Cellular basis of physiological and pathological myocardial growth. In:Page, E.; Fozzard, HA.; Solaro, RJ., editors. Handbook of Physiology: The CardiovascularSystem: The Heart. Oxford University Press; 2002. p. 75-144.

Goichberg et al. Page 11

Circ Res. Author manuscript; available in PMC 2012 April 29.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 12: NIH Public Access Domenico D’Amario João Ferreira … Circ Res. 2011.pdf · qRT-PCR and Western Blotting ... mice were injected with hCSCs pre-treated with ephrin A1 or Fc for

25. Cheng W, Reiss K, Li P, Chun MJ, Kajstura J, Olivetti G, Anversa P. Aging does not affect theactivation of the myocyte insulin-like growth factor-1 autocrine system after infarction andventricular failure in Fischer 344 rats. Circ Res. 1996; 78:536–546. [PubMed: 8635210]

26. Ueda H, Nakamura T, Matsumoto K, Sawa Y, Matsuda H, Nakamura T. A potentialcardioprotective role of hepatocyte growth factor in myocardial infarction in rats. Cardiovasc Res.2001; 51:41–50. [PubMed: 11399246]

27. Tillmanns J, Rota M, Hosoda T, Misao Y, Esposito G, Gonzalez A, Vitale S, Parolin C, Yasuzawa-Amano S, Muraski J, De Angelis A, Lecapitaine N, Siggins RW, Loredo M, Bearzi C, Bolli R,Urbanek K, Leri A, Kajstura J, Anversa P. Formation of large coronary arteries by cardiacprogenitor cells. Proc Natl Acad Sci USA. 2008; 105(5):1668–1673. [PubMed: 18216245]

28. Yamashita T, Ohneda K, Nagano M, Miyoshi C, Kaneko N, Miwa Y, Yamamoto M, Ohneda O,Fujii-Kuriyama Y. Hypoxia-inducible transcription factor-2alpha in endothelial cells regulatestumor neovascularization through activation of ephrin A1. J Biol Chem. 2008; 283:18926–18936.[PubMed: 18434321]

29. Kikuchi K, Holdway JE, Werdich AA, Anderson RM, Fang Y, Egnaczyk GF, Evans T, MacraeCA, Stainier DY, Poss KD. Primary contribution to zebrafish heart regeneration by gata4(+)cardiomyocytes. Nature. 2010; 464:601–605. [PubMed: 20336144]

30. Miao H, Li DQ, Mukherjee A, Guo H, Petty A, Cutter J, Basilion JP, Sedor J, Wu J, Danielpour D,Sloan AE, Cohen ML, Wang B. EphA2 mediates ligand-dependent inhibition and ligand-independent promotion of cell migration and invasion via a reciprocal regulatory loop with Akt.Cancer Cell. 2009; 16:9–20. [PubMed: 19573808]

31. Holmberg J, Armulik A, Senti KA, Edoff K, Spalding K, Momma S, Cassidy R, Flanagan JG,Frisen J. Ephrin-A2 reverse signaling negatively regulates neural progenitor proliferation andneurogenesis. Genes Dev. 2005; 19:462–471. [PubMed: 15713841]

32. Genander M, Holmberg J, Frisen J. Ephrins negatively regulate cell proliferation in the epidermisand hair follicle. Stem Cells. 2010; 28:1196–1205. [PubMed: 20506314]

33. Salaita K, Nair PM, Petit RS, Neve RM, Das D, Gray JW, Groves JT. Restriction of receptormovement alters cellular response: physical force sensing by EphA2. Science. 2010; 327:1380–1385. [PubMed: 20223987]

Goichberg et al. Page 12

Circ Res. Author manuscript; available in PMC 2012 April 29.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 13: NIH Public Access Domenico D’Amario João Ferreira … Circ Res. 2011.pdf · qRT-PCR and Western Blotting ... mice were injected with hCSCs pre-treated with ephrin A1 or Fc for

Figure 1. Expression of EphA2 and ephrin A1A, Transcripts for EphA2 and ephrin A1 in hCSCs, human cardiomyocytes (hCMs), mCSCsand mouse cardiomyocytes (mCMs). Representative tracings and quantitative data areshown. *P<0.05 vs. hCSCs or mCSCs. hMyo, human myocardium. For PCR products, seeOnline Figure I. B, Western blotting of EphA2 and ephrin A1 in hCSCs and hCMs. EphA2is restricted to hCSCs and ephrin A1 to hCMs. Loading, GAPDH or β-actin. C, hCMs (α-sarcomeric actin: α-SA, red) express ephrin A1 (white) and are negative for EphA2. DAPI:nuclei, blue. D, c-kit-positive hCSCs (left, green) express EphA2 on the plasma membrane(central, red). Right, merge. E, Five hCSCs (c-kit, green) are nested within the hMyo(hCMs: α-SA, red) and express connexin 43 (C×43, white) and N-cadherin (N-cadh,yellow). The area included in the rectangle is shown at higher magnification in the inset.Fibronectin, magenta. F, Ephrin A1 (white) is present in hCMs (α-SA, red) located inproximity of c-kit-positive hCSCs (green) expressing EphA2 (yellow). The area included inthe rectangle is shown at higher magnification in the inset. G, Ephrin A1 is restricted tohCMs and is not present in ECs (von Willebrand factor: vWf, magenta), SMCs (α-smoothmuscle actin: α-SMA, green) and fibroblasts (procollagen1α: procoll, light blue).

Goichberg et al. Page 13

Circ Res. Author manuscript; available in PMC 2012 April 29.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 14: NIH Public Access Domenico D’Amario João Ferreira … Circ Res. 2011.pdf · qRT-PCR and Western Blotting ... mice were injected with hCSCs pre-treated with ephrin A1 or Fc for

Goichberg et al. Page 14

Circ Res. Author manuscript; available in PMC 2012 April 29.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 15: NIH Public Access Domenico D’Amario João Ferreira … Circ Res. 2011.pdf · qRT-PCR and Western Blotting ... mice were injected with hCSCs pre-treated with ephrin A1 or Fc for

Figure 2. Ephrin A1 activation of hCSCsA, Transcripts for EphA2, EphA3 and EphA4 (left). siC, non-targeting siRNA. *P<0.05 vs.siC. Expression of EphA2 (right, red) in hCSCs transfected with siEphA2 or siC. Phalloidin,green. B, Fraction of hCSCs attached to immobilized ephrin A1 in the presence of non-targeting RNA (siC). Transfection of hCSCs with siRNA for EphA2 (siEphA2) abrogatedthe adhesion response. Human IgG coating (Fc) was used as control. *,**P<0.05 vs. Fc andsiC, respectively. C, Actin filaments (phalloidin, green) in hCSCs exposed to Fc or ephrinA1. Actin accumulates at the leading lamella of ephrin A1-activated hCSCs. D, hCSCsexpressing EphA2 (red) were exposed to Fc (upper panels) or ephrin A1 (lower panels).Internalization of the ephrin A1-EphA2 complex (red-white dots) occurs in ephrin A1-activated hCSCs. EphA2 is confined to the plasma membrane in Fc-exposed hCSCs. Fcalone and ephrin A1-Fc (white) were recognized by an antibody against the Fc portion ofhuman IgG. Phalloidin, green. E, Immunoblotting and quantitative analysis of Src familykinase phosphorylation in hCSCs exposed to ephrin A1 for 0, 5, 15 and 30 minutes (min).*P<0.05 vs. time 0. F, Directional stimulation of hCSCs with HGF results in EphA2 (red)accumulation at the leading edge (arrowheads) of the migrating cells. Actin filaments areshown by phalloidin (green). Arrow, direction of migration; c-kit, white. Control, hCSCs inthe absence of HGF. G, Number of migrated hCSCs. *P<0.05 vs. Fc. H, hCSCs migration isnegatively affected by EphA2 siRNA (siEphA2) and chemical inhibition of Src kinase. siC,non-targeting RNA. *P<0.05 vs. control conditions. I, mRNA for α-SA, α-SMA and vWf(left), and fraction of myocytes, SMCs and ECs formed by hCSCs in the presence andabsence of ephrin A1 (right). Control, hCSCs not exposed to dexamethasone.

Goichberg et al. Page 15

Circ Res. Author manuscript; available in PMC 2012 April 29.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 16: NIH Public Access Domenico D’Amario João Ferreira … Circ Res. 2011.pdf · qRT-PCR and Western Blotting ... mice were injected with hCSCs pre-treated with ephrin A1 or Fc for

Figure 3. Expression of ephrin A1 in diseased heartsA, Two days after myocardial infarction, the expression of ephrin A1 (white) increases inthe spared mCMs (right: α-SA, red). Sham-operated, SO (left). B, Immunoblotting of ephrinA1 and Src family kinase phosphorylation in myocardium obtained from SO, and from theremote region and border zone of infarcted mice. *P<0.05 vs. SO. C, Ephrin A1 mRNA inisolated hCMs from donor and explanted human hearts.

Goichberg et al. Page 16

Circ Res. Author manuscript; available in PMC 2012 April 29.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 17: NIH Public Access Domenico D’Amario João Ferreira … Circ Res. 2011.pdf · qRT-PCR and Western Blotting ... mice were injected with hCSCs pre-treated with ephrin A1 or Fc for

Figure 4. Ephrin A1 and mCSC migrationA, Two days after infarction, EGFP-positive-c-kit-positive (green) mCSCs are located at theborder zone of infarcted mice injected with Fc or ephrin A1. Low and high magnificationsare shown. mCMs: α-SA, red. MI, myocardial infarction. B, Number of mCSCs in theborder zone of Fc- and ephrin A1-treated infarcted mice. *P<0.05 vs. Fc. C, Fraction ofreplicating (Ki67-positive) and dying (TdT-positive) mCSCs in the same region.

Goichberg et al. Page 17

Circ Res. Author manuscript; available in PMC 2012 April 29.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 18: NIH Public Access Domenico D’Amario João Ferreira … Circ Res. 2011.pdf · qRT-PCR and Western Blotting ... mice were injected with hCSCs pre-treated with ephrin A1 or Fc for

Figure 5. Ephrin A1 and migration of hCSCs in the infarcted heartA, Images of the same field of the border zone of an infarcted heart are shown by two-photon-microscopy two hours after coronary artery ligation and injection of hCSCs. Upperpanels: infarcted heart injected with hCSCs exposed to Fc. Over a period of 80 min, clustersof EGFP-positive hCSCs (green) are confined to the site of injection (arrowheads). Lowerpanels: infarcted heart injected with ephrin A1-pretreated hCSCs. Clusters of EGFP-positivehCSCs moved in the direction of the arrow over a period of 80 min. B, Solid lines reflect thedisplacement per hour of individual hCSCs exposed to Fc or ephrin A1. Different colorscorrespond to different cells. Areas included in the rectangles are shown at highermagnification in the lower panels. C, Speed of migration of hCSCs within the myocardium.*P<0.05 vs. Fc.

Goichberg et al. Page 18

Circ Res. Author manuscript; available in PMC 2012 April 29.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 19: NIH Public Access Domenico D’Amario João Ferreira … Circ Res. 2011.pdf · qRT-PCR and Western Blotting ... mice were injected with hCSCs pre-treated with ephrin A1 or Fc for

Goichberg et al. Page 19

Circ Res. Author manuscript; available in PMC 2012 April 29.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 20: NIH Public Access Domenico D’Amario João Ferreira … Circ Res. 2011.pdf · qRT-PCR and Western Blotting ... mice were injected with hCSCs pre-treated with ephrin A1 or Fc for

Goichberg et al. Page 20

Circ Res. Author manuscript; available in PMC 2012 April 29.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 21: NIH Public Access Domenico D’Amario João Ferreira … Circ Res. 2011.pdf · qRT-PCR and Western Blotting ... mice were injected with hCSCs pre-treated with ephrin A1 or Fc for

Figure 6. Ephrin A1 and myocardial regenerationA, Myocardial scarring after infarction (collagen, white). A thin layer of spared myocytes(α-SA, red) is present in the subendocardium. The area included in the rectangle is shown athigher magnification in the inset. B and C, Band of regenerated myocardium after infarctionand delivery of EGFP-labeled hCSCs exposed to Fc (B) or ephrin A1 (C); EGFP and α-SA(yellowish). The areas included in the rectangles are shown at higher magnification in theinsets. D, Newly formed capillaries are positive for EGFP (green) and vWf (white). SMCs inregenerated arterioles are positive for EGFP and α-SMA (orange), and ECs for EGFP andvWf (white-green). E, hCMs are positive for EGFP (left, green) and Alu (left, white dots innuclei), and EGFP and α-SA (right, orange). F, hCMs carry the Y-chromosome (left, Y-chr,single white dot in nuclei), and are EGFP and α-SA positive (right). G and H, Spectralanalysis of hCMs positive for EGFP (G) and Alu (H). Emission spectra for EGFP (G) andAlu (H) positive hCMs are shown by the green lines, while emission spectra for tissueautofluorescence are shown by the blue lines. Note the difference in the intensity of thesignals at wavelength corresponding to maximum fluorescence in each case (upper panels).Following normalization for the intensity of the signals, all emission spectra for specificlabeling were essentially superimposable (lower panels). In contrast, emission spectra fortissue autofluorescence had different shapes and were easily distinguishable from specificlabeling.

Goichberg et al. Page 21

Circ Res. Author manuscript; available in PMC 2012 April 29.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 22: NIH Public Access Domenico D’Amario João Ferreira … Circ Res. 2011.pdf · qRT-PCR and Western Blotting ... mice were injected with hCSCs pre-treated with ephrin A1 or Fc for

Figure 7. Ephrin A1-EphA2 system and cardiac repairA, Number of myocytes lost and remaining after infarction in the LV of animals injectedwith PBS (MI), Fc-treated hCSCs (Fc) and ephrin A1-activated (ephrin A1) hCSCs.*P<0.05 vs. SO. B, Characteristics of myocytes formed by differentiation of hCSCs inhearts injected with Fc-treated and ephrin A1-treated hCSCs. C and D, Length of newly-formed resistance arterioles (C) and capillaries (D) in the regenerated myocardium afterinjection with Fc-or ephrin A1-treated hCSCs. E, Reduction of infarct size by tissueregeneration. L: lost myocardium; R: regenerated myocardium. *P<0.05 vs. Fc.

Goichberg et al. Page 22

Circ Res. Author manuscript; available in PMC 2012 April 29.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 23: NIH Public Access Domenico D’Amario João Ferreira … Circ Res. 2011.pdf · qRT-PCR and Western Blotting ... mice were injected with hCSCs pre-treated with ephrin A1 or Fc for

Figure 8. Ephrin A1 and functional recoveryA, Hemodynamics and echocardiographic parameters in SO, and in infarcted hearts injectedwith PBS (MI), Fc-treated hCSCs (Fc), and ephrin A1-treated hCSCs (ephrin A1).*,**,†P<0.05 vs. SO, MI, and Fc, respectively. B, Number of arrhythmic events in SO, andinfarcted hearts injected with PBS (MI), Fc-treated hCSCs (Fc), and ephrin A1-treatedhCSCs (ephrin A1). *,**P<0.05 vs. SO and MI, respectively.

Goichberg et al. Page 23

Circ Res. Author manuscript; available in PMC 2012 April 29.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript