Está en la página 1de 15

STEM CELLS AND DEVELOPMENT ORIGINAL RESEARCH REPORT

Volume 00, Number 00, 2014


 Mary Ann Liebert, Inc.
DOI: 10.1089/scd.2013.0305

Stable Phenotype and Function of Immortalized


Lin Sca-1 + Cardiac Progenitor Cells in Long-Term Culture:
-

A Step Closer to Standardization

Ana G. Freire,1,2,* Diana S. Nascimento,1,* Giancarlo Forte,3,4,* Mariana Valente,1,5 Tatiana P. Resende,1
Stefania Pagliari,3,4 Cláudia Abreu,6 Isabel Carvalho,7 Paolo Di Nardo,3 and Perpétua Pinto-do-Ó1,5

Putative cardiac progenitor cells (CPCs) have been identified in the myocardium and are regarded as promising
candidates for cardiac cell-based therapies. Although two distinct populations of CPCs reached the clinical setting,
more detailed studies are required to portray the optimal cell type and therapeutic setting to drive robust cell
engraftment and cardiomyogenesis after injury. Owing to the scarcity of the CPCs and the need for reproduc-
ibility, the generation of faithful cellular models would facilitate this scrutiny. Here, we evaluate whether im-
mortalized Lin - Sca-1 + CPCs (iCPCSca-1) represent their native-cell counterpart, thereby constituting a robust in
vitro model system for standardized investigation in the cardiac field. iCPCSca-1 were established in vitro as plastic
adherent cells endowed with robust self-renewal capacity while preserving a stable phenotype in long-term
culture. iCPCSca-1 differentiated into cardiomyocytic-, endothelial-, and smooth muscle-like cells when subjected
to appropriate stimuli. The cell line consistently displayed features of Lin - Sca-1 + CPCs in vitro, as well as
in vivo after intramyocardial delivery in the onset of myocardial infarction (MI). Transplanted iCPCSca-1 sig-
nificantly attenuated the functional and anatomical alterations caused by MI while promoting neovascularization.
iCPCSca-1 are further shown to engraft, establish functional connections, and differentiate in loco into cardio-
myocyte- and vasculature-like cells. These data validate iCPCSca-1 as an in vitro model system for Lin - Sca-1 +
progenitors and for systematic dissection of mechanisms underlying CPC subsets engraftment/differentiation
in vivo. Moreover, iCPCSca-1 can be regarded as a ready-to-use CPCs source for pre-clinical bioengineering
studies toward the development of novel strategies for restoration of the damaged myocardium.

Introduction fects have been reported, the improvement of cardiac


function has been greatly ascribed to the release of paracrine

C ardiac cell therapy was shown to be a very complex


endeavor for which a completely innovative vision and
novel technologies are necessary. Indeed, in spite of intensive
factors rather than to an increased presence of functional
cardiomyocytes (CM) in the injured myocardium [3–5].
Regardless, in the last decade, adult cardiac progenitor cells
scientific and economic efforts, the technological exploita- (CPCs) have gathered abundant attention [6–18], paving the
tion of the stem cell potential has generated frustrating results way to a quick transition from the bench to the bedside
owing to the unsystematic and simplistic approaches con- [19,20]. In fact, two independent clinical trials, each ad-
sidered so far. Myocardial cell delivery in experimental an- dressing a different subset of progenitor cells (c-Kit-
imals and patients results in a very low number (*3–10%) of expressing cells for SCIPIO [19] and cardiosphere-derived
cell persistence in the myocardium due to significant cell cells for CADUCEUS [20]), were carried out to assess their
death and/or loss to the bloodstream with subsequent en- therapeutic potential in the myocardial infarction (MI)
trapment into the lungs [1,2]. Although some beneficial ef- setting. Although the short-term results on a reduced patient

1
INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.
2
Faculdade de Engenharia da Universidade do Porto, Universidade do Porto, Porto, Portugal.
3
Laboratorio di Cardiologia Molecolare and Cellulare, Dip. di Medicina Interna, Università di Roma Tor Vergata, Rome, Italy.
4
International Clinical Research Center (ICRC), Integrated Center of Cellular Therapy and Regenerative Medicine, St. Anne’s University
Hospital, Brno, Czech Republic.
5
Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal.
6
Cardiologia Veterinária Móvel, Universidade do Porto, Porto, Portugal.
7
IBMC—Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.
*These three authors contributed equally to this work.

1
2 FREIRE ET AL.

number have shown a partial amelioration of the heart Methods


condition, most aspects of the CPCs biology are still un-
Animals
defined. Hence, only an indisputable demonstration of the
beneficial effects of these strategies can suppress the innu- C57BL/6 mice aged 6–12 weeks were used. Procedures
merable doubts regarding protocols based on harsh manip- were approved by Instituto de Biologia Molecular e Ce-
ulations of stem/progenitor cells [21]. Among many others, lular–Instituto de Engenharia Biomédica Animal Ethics
the factors governing the admirable equilibrium of the Committee and National Direção Geral de Veterinária
myocardial in vivo complexity, such as the interactions (permit no: 022793), and are in conformity with the Euro-
between progenitor cells and environmental components, pean Parliament Directive 2010/63/EU. Humane endpoints
remain largely undefined. Therefore, the only possibility to were followed according to the Organization for Economic
mimic the dynamics of myocardial homeostasis in order to Co-operation and Development Guidance Document on the
establish safe and reliable repair procedures is a stringent Recognition, Assessment, and Use of Clinical Signs as
‘‘standardization’’ of the different aspects of myocardial Humane Endpoints for Experimental Animals Used in
complexity [22]. Safety Evaluation (2000).
A clear understanding on CPCs biology is impaired by Mice were anesthetized by an intraperitoneal injection
the scarcity of progenitors present within the myocardium (ip) of medetomidine (1 mg/kg; Sededorm) and ketamine
as well as by time-consuming isolation procedures, and, (75 mg/kg; Clorketam), and its adequacy was monitored by
importantly, by their phenotypic variability when isolated the pedal withdrawal reflex.
and expanded in different laboratories [23]. Consistently,
progenitor cells that apparently partake in the same pop-
Isolation and culture of Sca-1 + CPCs
ulation can display non-comparable features. To circum-
vent these limitations, the aim of this study was to create a CPCs were isolated as previously described [29]: 6 week
benchmark to be used as a reference for progenitor cell male C57BL/6 mice hearts were minced and digested us-
populations isolated from the myocardium in different ing 0.25% Trypsin/ethylenediaminetetraaceticacid (EDTA)
laboratories on the basis of a plethora of non-specific cell- and collagenase II (1,500 U; Worthington) in phosphate-
surface markers [6–15,17,18]. For this purpose, lineage buffered saline (PBS). Tissue fragments were cultured in
negative/Sca-1 + (Lin - Sca-1 + ) CPCs have been isolated Dulbecco’s Modified Eagle Medium (DMEM; Lonza) that
from murine hearts and immortalized via telomerase cat- was supplemented with 10% fetal calf serum (Lonza),
alytic subunit [murine telomerase catalytic subunit 100 IU/mL penicillin and 100 mg/mL streptomycin, insulin-
(mTERT)] overexpression. Recently, the role of telomer- transferrin-selenium 1 · , 300 ng/mL retinoic acid, 0.8 mg/mL
ase activity in maintaining CPC viability and regenerative linoleic acid, 2 mM l-glutamine, 0.1 ng/mL insulin-like growth
potential was demonstrated [24]. Aging leads to telomeric factor 1, and 0.1 ng/mL endothelial growth factor (complete
shortening in CPCs, thus leading to a senescent phenotype, medium). After 12–15 days, adherent cells migrating out
as shown by the expression of p16INK4a. Such events have from fragments were harvested and magnetically sorted
been correlated to cardiac function impairment, suggesting using anti-hematopoietic lineages antibody cocktail (Mil-
that CPC loss could be a main determinant in heart failure. tenyi Biotec). Sca-1 + cells were enriched from the Lin -
Moreover, the overexpression of the human telomerase fraction by two sets of magnetic cell sorting protocols
reverse transcriptase (TERT) catalytic subunit in various (Miltenyi Biotec) and expanded in culture in complete
cell lines resulted in extended cellular lifespan [25–27] medium before transfection.
without detectable changes characteristic of malignant
transformation [28]. Establishment of iCPCSca-1
In the present study, a cell line of immortalized Sca-1 +
cardiac progenitor cells (iCPCSca-1) has been specifically Murine TERT coding sequence, obtained from pGRN190
generated for the first time. iCPCSca-1 were extensively (kind gift from Geron Corporation), was inserted into EcoRI
characterized to assess whether after immortalization these restriction site in pcINeo plasmid (Promega). Lin - Sca-1 +
cells had preserved the hallmarks of their native-cell coun- cells were transfected with pCINeo-TERT or control vector
terpart and the recognized capability to engraft and differ- using the Calcium Phosphate Method (Promega), according
entiate when transplanted in an MI murine model. Cells to the manufacturer’s instructions and incubated in G418-
grew in vitro as adherent cells with a typical spindle-shape containing medium (Sigma-Aldrich) for at least 3 weeks.
morphology and displayed robust self-renewal capacity, For clonal selection, immortalized cells were plated at
while preserving a CPC tri-lineage potential (ie, differenti- single-cell density by limiting dilution and cultured in
ated in CM-, endothelial-, and smooth muscle like-cells). Sca-1 + -conditioned medium. Clones derived from a single
iCPCSca-1 transplanted into the MI border zone significantly cell were expanded under G418 selection and character-
reduced the MI-induced left ventricle (LV) anatomical and ized for Sca-1 expression by flow cytometry. Proliferative
functional alterations. Importantly, the cells engrafted, es- capacity of the established line was calculated using the
tablished functional connections, and differentiated in loco formula log10 (total no./start no.)/log2.
into cardiomyocyte- and vasculature-like cells, as previously
shown for their native counterparts. Thus, the iCPCSca-1 line In vivo tumorigenicity assay
constitutes an unlimited source of Sca-1 + CPC replicates
and can be used as a model system for in vitro high- The tumorigenicity of iCPCSca-1 was assayed by a het-
throughput studies and for the dissection of the in vivo role erotopic injection into syngeneic animals [30,31]. Eight-
of adult heart-resident progenitors. week-old C57BL/6 female mice were subjected to a
SCA-1
ICPC —A BENCHMARK FOR HEART SCA-11 PROGENITORS 3

subcutaneous injection of 1 · 106 cells [in 0.2 mL of 2% days [32], or (iii) maintained using a commercial cardio-
fetal bovine serum (FBS) in a-minimum essential medium myocyte differentiation medium (Millipore) for 21 days.
(a-MEM)] of iCPCSca-1 at each shoulder pad. Animals were For the co-culture set-up, CM were isolated from neonatal
monitored daily for the appearance of palpable tumors. Mice (postnatal day 1–3) C57BL/6 as previously described [33].
were necropsied after a 5-month surveillance and the in- iCPCSca-1 were pre-stained with viable red fluorescent dye
jection region as well as the heart, spleen, lung, and liver VybrantTM DiI (Molecular Probes) before seeding onto
were harvested, fixed in 10% formalin neutral buffer (VWR CMs for 7 days.
BDH and Prolabo), and paraffin embedded. For endothelial differentiation, iCPCSca-1 were pre-
conditioned for 10 days in Endothelial Growth Medium with
Flow cytometry EGM BulletKit (EGM; Lonza) or in a-MEM (Gibco) with
10% FBS as control. After this period, cells were plated for
Before flow-cytometry analysis, single-cell suspensions immunocytochemistry (ICC), processed for CD31 flow-
were labeled with the following antibodies: APC—anti- cytometry staining, or seeded onto Matrigel Growth Factor-
PDGFRa (Biolegend), c-Kit and Flk-1 (eBiosciences); PE— Reduced Matrix (BD Biosciences). After overnight culture,
anti-CD105 (Biolegend), CD106 (Biolegend), CD44 (Im- cells were fixed and stained for vWF (PBS/0.05% Tween-
munotools), CD45 (Immunotools), CD34 (Biolegend), 20/1% BSA for 4 h at RT) or for calcein AM (BD Bios-
CD31 (BD Pharmingen), CD62L (Immunotools), CD40L ciences). Briefly, cells were incubated for 30 min at 37C in
(BD Pharmingen), CD90.2 (BD Pharmingen), and Stro-1 calcein/PBS and imaged using the inverted fluorescence
(Santa Cruz Biotech); and FITC—anti Sca-1 (eBiosciences) microscope Axiovert 200 (Zeiss). To evaluate tube forma-
and CD40 (BD Pharmingen). Samples were acquired on an tion, number of branch points and tube length were assessed
FACS Canto II and analyzed using FlowJo software. using ImageJ1.42.
For smooth muscle cell differentiation, iCPCSca-1 were
Gene expression cultured in DMEM high glucose containing 2% FBS and
50 ng/mL of platelet-derived growth factor BB (PDGF-BB;
For the reverse transcription polymerase chain reaction
R&D Systems) for 10 days, as described earlier [7].
(RT-PCR), total RNA was extracted using Trizol Reagent
(Sigma-Aldrich), reverse transcribed according to the man-
ufacturer’s instructions (Bioline) and PCR was performed MI, iCPCSca-1 delivery, and echocardiography
using BIOTAQ DNA polymerase (Bioline) and gene-specific
MI was induced by permanent ligation of left anterior
primers. For quantitative RT-PCR (qRT-PCR) analysis,
descending (LAD) coronary artery as described elsewhere
cDNAs were synthesized using PrimeScriptTM RT reagent
[34] with minor alterations. After anesthesia, female
kit following the manufacturer’s instructions (Takara Bio,
C57BL/6 were intubated and ventilated using a small-
Inc.). qRT-PCR was performed using iQTM Sybr Green
animal respirator (Minivent 845; Harvard Apparatus). The
Supermix (Bio-Rad) and gene-specific primers. Reactions
heart was exposed (Ø 5–7 mm) via left thoracotomy on the
were carried out in triplicate on the iCycler iQ5 Real-Time
third intercostal space, and the pericardial sac was gently
PCR system (Bio-Rad). Values were normalized to glycer-
disrupted. A non-absorbable 7-0 suture (Silkam; B. Braun)
aldehyde-3-phosphate dehydrogenase housekeeping gene.
was used for ligation. Immediately afterward, 5 · 105
Primer sequences are available on request.
iCPCSca-1 (n = 8) or vehicle (0.5% BSA/PBS) only (n = 9)
were delivered by four intramyocardial injections (5 mL
Immunocytochemistry each) with a 30-gauge syringe (Hamilton company). Thor-
iCPCSca-1 were fixed with 4% paraformaldehyde (PFA) or acic incision was closed using an absorbable 6-0 suture
cold methanol for GATA-4 detection, permeabilized with (Safil; B. Braun), and surgical staples were used for skin
0.2% (cytoplasmic) or 1% Triton X-100/PBS (nuclear epi- closure. Anesthesia was reverted by atipamezole (ip, 5 mg/
topes), and blocked for 1 h with 1% goat serum, 1% bovine kg; Revertor). Analgesia and fluid therapy were performed
serum albumin (BSA), or M.O.M. Immunodetection Kit by ip delivery of butorphanol (1 mg/kg; Butador) and a
(Vector Lab). Incubation with anti-Nestin (Abcam), anti- subcutaneous injection of 5% glucose physiological saline,
GATA-4 (Santa Cruz Biotech.), anti-Sarcomeric a-actinin (a- respectively. This procedure was repeated every 12 h for
actinin; Sigma-Aldrich), anti-a smooth muscle actin (a-SMA; approximately 72 h post-surgery or until full recovery.
Sigma-Aldrich), or anti-von Willebrand Factor (vWF; Sigma-
Aldrich) was carried out for 1–2 h at room temperature (RT). Echocardiographic assessment
Incubation with secondary antibodies was for 1 h at RT, with
the exception of M.O.M. Biotinylated Anti-Mouse IgG Transthoracic echocardiography was performed at 2
Reagent (Vector Lab) by which the manufacturer’s instruc- weeks after LAD coronary artery ligation using a portable
tions were followed. Slides were mounted in Vectashield with ultrasound apparatus (GE Vivid I; General Electric) that was
4,6-diamino-2-phenylindole (DAPI; Vector Lab) and observed equipped with a 12 MHz linear probe (GE 12L-RS Linear
on the inverted fluorescence microscope Axiovert 200 (Zeiss). Array Transducer; General Electric). To evaluate LV
structural changes, several parameters from M-mode were
iCPCSca-1 differentiation measured, that is, the LV internal diameter at diastole
(LVIDd) and at systole (LVIDs), the interventricular septum
For cardiomyogenic differentiation, cells were (i) co- at diastole and at systole, the LV posterior wall at diastole
cultured with neonatal CM for 7 days, (ii) cultured in the and at systole (LVPWs), and heart rate. Left ventricular
presence of transforming growth factor-b (TGF-b) for 21 ejection fraction (EF) and fraction shortening (FS) were
4 FREIRE ET AL.

calculated as an index of systolic function: FS (%) = CD31 for 1–2 h at RT, 1 h incubated with secondary anti-
[(LVIDd - LVIDs)/LVIDd] ·100 and EF (%) = [(LVIDd3– body and mounted in Vectashield with DAPI. Images were
LVIDs3)/LVIDd3] ·100. captured using MosaiX (AxioVision modules; Carl Zeiss).
CD31 + cells were counted in 30 fields per heart over 3–5
Histological procedures sections at infarcted and border zone regions along the
heart’s long axis (n = 9 control group, n = 8 iCPCSca-1) using
Hearts were harvested at 2 weeks post-surgery after an ImageJ1.42. Density was calculated as CD31-positive cells
injection with potassium chloride and fixed in 10% Formalin per square millimeter (mm2).
neutral buffer (VWR BDH and Prolabo) for approximately
24 h before paraffin embedding. Representative LV sam- Data and statistical analysis
pling (*12 sections at 3 mm) was obtained by transverse
sectioning from apex to base with 300 mm intervals. Statistical significance was evaluated with SPSS v.19.0
Masson’s Trichrome (MT) staining was performed using using Mann–Whitney or one-way analysis of variance with
the Trichrome (Masson) Stain kit (Sigma-Aldrich) with the post hoc Tukey’s test. Values are presented as mean – stan-
following modifications: Nuclei were pre-stained with Ce- dard error of the mean. P < 0.05 was considered statistically
lestine Blue solution following Gill’s Hematoxylin staining, significant. *P < 0.05, **P < 0.01, and ***P < 0.001.
and tissue was incubated for 1 h in Bouin’s solution before
muscle staining with Biebrich Scarlet-Acid Fuchsin. Sec- Results
tions were photographed with a stereomicroscope (Leica)
before morphometric analysis. Generation of an immortalized line representative
of Lin - Sca-1 + adult CPCs
MI size calculation and morphometric analysis
To immortalize Lin - Sca-1 + CPCs by inducing ectopic
Infarct size measurement was based on collagen deposi- expression of mTERT, cells expressing Sca-1 were isolated
tion in the ischemic LV wall 2 weeks post-infarction and from murine hearts and transfected with pCINeo vector
calculated by the area method using the semi-automated encoding the mTERT.
program MIQuant [35]. The thickness of LV-free wall was Further analysis was restricted to one transfected clone
measured on the infarction region using ImageJ1.42 on MT- of myocardium-resident Sca-1 + progenitors, that is, the
stained sections. iCPCSca-1. The remaining G418-resistent isolated clones
were subjected to transcriptional profiling (Supplementary
Fluorescence in situ hybridization Table S1; Supplementary Data are available online at www
.liebertpub.com/scd) and cryopreserved for future investigation.
Sections were subjected to antigen retrieval using 10 mM iCPCSca-1 grew to confluence as a monolayer of spindle-
sodium citrate Buffer (pH 6; Sigma-Aldrich) at 98C for shaped adherent cells similar to their native counterparts (Fig.
10 min followed by 0.01% acid pepsin (Sigma-Aldrich) at 1A) and presented a doubling time of *21.6 h. iCPCSca-1 were
37C for 10 min. For fluorescence in situ hybridization continuously sub-cultured for more than 24 months with no
(FISH), samples were dehydrated and incubated with a spe- signs of differentiation and maintained high levels of Sca-1
cific probe to whole mouse Y-chromosome (Y-Chr; Cambio) expression. Conversely, primary (non-transfected) Lin - Sca-
for 5 min at 82C and overnight at 37C. Samples were in- 1 + CPC decreased Sca-1 expression after 1 month in culture
cubated with Streptavidin (Invitrogen) for 30 min, mounted in and senesced after 15–20 passages (Supplementary Fig. S1).
Vectashield with DAPI (Vector Lab), and observed on the Genome integration of mTERT was confirmed by
inverted fluorescence microscope Axiovert 200 (Zeiss). Southern blotting using a specific probe for the neomycin
For a-actinin and CD31 (Santa Cruz Biotech.) im- phosphotransferase gene (Fig. 1B). Chromosome number
munostaining combined with FISH (immuno-FISH), antigen per metaphase plate of iCPCSca-1 was compared with Sca-
retrieval was achieved by pronase treatment for 30 min at 37C 1 + primary cells; the majority of the cells were diploid
or with 10 mM Tris/1 mM EDTA (pH 9.0) at 98C for 30 min. and displayed a median chromosome number of 44 and 40
Samples were permeabilized with 0.2% Triton X-100 and in iCPCSca-1 and primary culture, respectively (Fig. 1C). In
blocked using 4% FBS and 1% BSA or M.O.M Im- addition, tetraploid cells were present in both conditions.
munodetection Kit (Vector Lab). Incubation with anti- The tumorigenic potential of iCPCSca-1 was evaluated by
Connexin43 (Cx43; Abcam), anti-a-SMA(Sigma-Aldrich) means of a classical tumorigenesis assay in which syngeneic
and anti-CD31 was performed at RT for 1–2 h and overnight at animals are used as recipients for heterotopic delivery of the
4C for anti-a-actinin (Sigma-Aldrich). After a 1 h incubation cell inocula [30]. Hence, iCPCSca-1 were subcutaneously
with the secondary antibody (Invitrogen), slides were rinsed in delivered to the shoulder pads of syngeneic animals that
50 mM MgCl2/PBS for 5 min and post-fixed with 4% PFA and were daily monitored for tumor progression. No local mass
50 mM MgCl2/PBS for 10 minutes. Samples were dehydrated formation was observed after a 5-month inspection period
and incubated with Y-Chr probe as described earlier. and normal histological appearance was verified for the
heart, spleen, lungs, and liver (Supplementary Fig. S2).
Blood vessel density quantification
iCPCSca-1 display a typical tri-lineage potential
After antigen retrieval using 10 mM Tris/1 mM EDTA and a stable phenotype after long-term in vitro culture
(pH 9.0; Sigma-Aldrich) at 98C for 30 min, samples were
permeabilized with 0.2% Triton X-100 and blocked for 1 h Prospective identification of bona fide CPCs based on the
with 4% FBS and 1% BSA. Slides were incubated with anti- unique expression of surface markers is at the present not
FIG. 1. Generation and characterization of immortalized Lin - Sca-1 + cardiac progenitors (iCPCSca-1) cell line. (A) Rep-
resentative image of primary (CPCSca-1) and immortalized (iCPCSca-1) Sca-1 + CPC. (B) Successful integration of pCINeo-murine
telomerase catalytic subunit into iCPCSca-1 genome was confirmed by Southern blotting for Neomycin. The empty plasmid was
used as a positive control. (C) Chromosome counts on (-) iCPCSca-1 metaphase spreads show aneuploidy (n = 42) when
compared with (-) Sca-1 + counterparts (n = 23). (D) Flow cytometry analysis of iCPCSca-1 displays high expression of Sca-1 and
mesenchymal-associated antigens (PDGFRa, CD29, CD44, CD105, and CD106). iCPCSca-1 lack hematopoietic- (CD45, c-kit)
and endothelial-affiliated (Flk-1, CD31, and CD34) markers. Bars are represented as mean – standard error of the mean (SEM).
(E). Reverse transcription polymerase chain reaction (RT-PCR) profile of primary CPCSca-1 and iCPCSca-1 reveals the expression
of stemness-related genes (Bcrp1, Bmi-1, and Nestin) and early cardiac transcriptional regulators (Gata-4, Isl-1, Mef2c, and Tef-
1). Mature contractile myofilaments [a- and b-cardiac myosin heavy chain beta (Mhc)] and pluripotency (Sox2, Nanog, and Oct4)
transcripts were not detected. Embryonic stem cells (ESC); bone marrow (BM), and heart (Hrt) were used as controls. (F) Flow
cytometry displays the profile of Sca-1 expression in primary CPCSca-1 and iCPCSca-1, and immunocytochemistry (ICC) cor-
roborates the expected sub-cellular location of progenitor-associated proteins GATA-4 and Nestin. Scale bar: 10 mm. Color
images available online at www.liebertpub.com/scd
5
6 FREIRE ET AL.

possible. Hence, it is generally accepted that an accredited cells, as a result of clone selection. Flow cytometry analysis
CPC will meet the following criteria: (i) derive from the clearly exposed the enrichment for the Sca-1-expressing
adult heart; (ii) display either Sca-1 or c-Kit at their surface fraction in CPCSca-1 (91.8%) and iCPCSca-1 (99.8%).
and lack markers of hematopoietic and endothelial com- Moreover, likely as a result of clonal selection, the iCPCSca-1
mitment/differentiation; (iii) express cardiac-affiliated tran- displayed a stronger and less disperse pattern of Sca-1 ex-
scription factors while lacking mature proteins; and (iv) pression as compared with primary cells. Proper sub-cellular
differentiate both in vitro and in vivo into CM, endothelial allocation of stemness-associated proteins GATA-4 and
cells, and smooth muscle cells. iCPCSca-1 were, thus, char- Nestin was further confirmed by ICC for both primary and
acterized with regard to the cell surface phenotype, tran- immortalized cells (Fig. 1F).
scriptional profile, and functional properties in order to To test iCPCSca-1 multipotency, distinct sub-passages
verify whether this cell line meets the premises mentioned were cultured under cell differentiation conditions. Cardio-
earlier. myocytic differentiation potential was evaluated by co-
Flow cytometry analysis indicated a phenotype consistent culture with mouse neonatal CM. iCPCSca-1 were pre-stained
with adult Lin - Sca-1 + CPCs, that is, a population consti- with viable red fluorescent dye Vybrant DiI to enable de-
tuted of cells with high Sca-1 levels (99.8% – 0.21%), tection in the co-culture system. After a week, iCPCSca-1 up-
lacking hematopoietic (Stro-1, CD45, CD34) and endothe- regulated GATA-4, displayed a functional pattern of Cx43
lial (Flk-1, CD31 and CD34) markers, and expressing and a typical myofibrillar assembly of sarcomeric a-actinin
mesenchymal-associated proteins (eg, CD29, CD44, (a-actinin) (Fig. 2A). Although the gold-standard protocol to
CD105, CD106, and PDGFRa), several of which are critical trigger CPCs differentiation is the direct co-culture with
to cell migration, adhesion, and communication (Fig. 1D). neonatal CM [33], this assay is prone to misleading inter-
Furthermore, the characterized clone (Clone No. 3, Sup- pretations due to spontaneous cell fusion events [38,39].
plementary Table S1) did not express c-Kit, a protein that Hence, we further addressed iCPCSca-1 differentiation in CM
singly or together with Sca-1 has been used to isolate CPCs. by culturing the cell line for 21 days in cardiomyocytic
A detailed transcriptional profile was carried out in parallel differentiation medium (Millipore) or in the presence of
with primary Sca-1 + CPC (CPCSca-1), embryonic stem cells, TGF-b. In this condition, iCPCSca-1 were also able to up-
bone marrow, and heart as controls. The side-by-side tran- regulate a-actinin expression when compared with cells
scriptional analysis demonstrated that iCPCSca-1 and pri- subjected to basal, that is, cell-maintenance, conditions for
mary Sca-1 + cells consistently expressed stemness-related the same time period (Fig. 2B). Furthermore, when iCPCSca-1
genes (Bcrp1, Bmi-1, and Nestin) and early cardiac tran- were transduced with a green fluorescent protein (GFP) re-
scription factors (Gata-4, Isl-1, Mef2c, and Tef-1) while porter under the cardiac troponin T (cTnT) promoter and
lacking transcripts for mature contractile myofilaments (a- subjected to TGF-b [32] for 45 days, approximately 97% of
and b-Mhc), fibroblasts [transcription factor 21 (Tcf21)], cells displayed GFP expression (Supplementary Fig. S3).
and regulators of pluripotency (Sox2, Nanog, Oct4), thus Since cTnT and a-actinin are also expressed in the devel-
corroborating that iCPCSca-1 preserves the phenotype of oping skeletal muscle, we evaluated the possibility of
cardiac-affiliated Sca-1 + multipotent progenitors (Fig. 1E). iCPCSca-1 having a skeletal origin. The latter was clearly
It should be noted that both primary and immortalized excluded by demonstrating the up-regulation of cardiac
CPCSca-1 expressed c-Myc, a transcription factor known to myosin heavy chain beta expression after differentiation,
block differentiation in distinct model systems [36] and while no detectable expression of myogenic differentiation
particularly shown to synergize with Pim-1 to induce the 1(MyoD), a marker of skeletal commitment, was found
proliferation and survival of CPCs [37]. Overall, the data (Fig. 2B). Endothelial differentiation potential was assessed
demonstrated that the immortalization of the cells did not by priming iCPCSca-1 with EGM medium for 10 days fol-
alter the transcriptional profile of Sca-1 + CPCs. The only lowed by a classical Matrigel assay to evaluate the capacity
obvious difference between the established line and the of the cells to form tubular-like structures. Primed iCPCSca-1
native counterparts is the lack of c-Kit in the immortalized seeded onto Matrigel exhibited tube-like interconnected


FIG. 2. iCPCSca-1 in vitro differentiation potential. (A) Immunostaining demonstrating an increase in GATA-4 expression
and presence of Cx43 and a-actinin in VybrantTM-stained iCPCSca-1 after 1 week of co-culture with neonatal cardiomyocytes.
Scale bar: 20 mm. (B) iCPCSca-1 a-actinin expression is increased after 21 days of culture in cardiomyocytic differentiation
conditions: transforming growth factor b or commercial medium (Millipore). Scale bar: 20 mm. RT-PCR representing the up-
regulation of b-MHC in iCPCSca-1 cultured in Millipore medium, while no MyoD transcript was observed in basal conditions
or after differentiation. (C) Calcein staining evidencing the tubular structures formed by human umbilical vein endothelial cells
(HUVECs) and iCPCSca-1 [pre-conditioned in either a-minimum essential medium (a-MEM) or endothelial growth medium
(EGM)] on Matrigel. Scale bar: 200 mm. The tube length and the number of branch points formed in each culture condition
were assessed. Data are represented as mean – SEM [n = 6, ***P < 0.001; **P < 0.01; *P < 0.05 and not significant (ns) by one-
way analysis of variance (ANOVA) with post hoc Tukey’s test]. EGM-preconditioned iCPCSca-1 grown on Matrigel exhibit
increased von Willebrand Factor (vWF) and CD31 expression as assessed by ICC and flow cytometry, respectively. Scale bars:
20 mm and 5 mm. Data are represented as mean – standard deviation (SD). (D) a-Smooth muscle actin (a-SMA) expression in
control iCPCSca-1 and after 10 days of culture in differentiation medium containing PDGF-BB. Scale bar: 20 mm. Histogram
plots show control in white and specific staining in black. Data are represented as mean – SD. qRT-PCR revealed an up-
regulation in a-SMA, SM22, and Cnn1 mRNA levels; while no dramatic alteration in Desmin expression levels was found. No
detectable levels of expression were observed for Myocd and Tcf21 (n.d.). Data are represented as a fold increase relative to
basal condition (n = 3). Color images available online at www.liebertpub.com/scd
SCA-1
ICPC —A BENCHMARK FOR HEART SCA-11 PROGENITORS 7
8 FREIRE ET AL.

structures, similar to the capillary assembly of human um- mRNA levels. In addition, no detectable expression of Myocd
bilical vein endothelial cells (HUVECs) (Fig. 2C, n = 6), was found, even on PDGF-BB stimulation. Since some of the
whereas unprimed iCPCSca-1 (iCPCSca-1 pre-cultured in a- proteins mentioned earlier are also displayed by fibroblasts/
MEM) were unable to assemble a fully organized capillary myofibroblasts, expression of the fibroblast marker Tcf21 was
network (n = 6). In fact, the tubular complex formed by un- evaluated in order to discard a fibroblastic origin of the
primed cells presented a significantly smaller number of iCPCSca-1. Detectable expression of Tcf21 was not found,
branch points (P < 0.001) and tube length (P < 0.05 and neither in basal conditions nor after PDGF-BB treatment (Figs.
P < 0.01) as compared with that assembled by EGM-primed 1E and 2D). Overall, these results demonstrate that iCPCSca-1
iCPCSca-1 and HUVECs (Fig. 2C). An endothelial phenotype were not derived from cardiac fibroblasts and that, after PDGF-
was further addressed by analyzing vWF and CD31 protein BB stimulation, cells undergo an incomplete smooth-muscle
expression. vWF was only detected in iCPCSca-1 cells after differentiation program as the levels of Myocd and Desmin
EGM priming (Fig. 2C). Accordingly, CD31 was increased have not changed after PDGF-BB treatment.
by 1.75% on primed cells when compared with iCPCSca-1 Taken together, the data demonstrate that iCPCSca-1 pre-
maintained in a-MEM for the same period (Fig. 2C). served the in vitro phenotype and functional ability to mod-
The capacity to differentiate into smooth muscle cells was erately differentiate as described by our team [29,33] and
evaluated by subjecting iCPCSca-1 to PDGF-BB for 10 days. others [5,12,13,16] for the native Lin - Sca-1 + counterparts.
After this period, iCPCSca-1 became elongated and displayed
a-SMA protein expression, as demonstrated by ICC and flow iCPCSca-1-transplanted hearts show improved
cytometry (27%) (Fig. 2D). In addition, the expression levels systolic function and lessened LV remodeling
of the smooth muscle-affiliated genes, a-SMA, Myocardin after MI
(Myocd), Transgelin (SM22), Calponin 1 (Cnn1), and Desmin
were also assessed by qRT-PCR (Fig. 2D). After 10 days of The beneficial effect of Lin - Sca-1 + CPCs administra-
PDGF-BB treatment, iCPCSca-1 up-regulated a-SMA, SM22, tion into the MI heart has been clearly established. No-
and Cnn1; while no clear alteration was observed for Desmin tably, CPCs are able to improve cardiac function and

FIG. 3. Functional and histological assessment of ischemic hearts transplanted with either iCPCSca-1 or vehicle. (A) Both
iCPCSca-1 and vehicle-injected groups display similar myocardial infarction (MI) size. (B) Ejection fraction and fractional
shortening measured in non-manipulated animals, vehicle-control and iCPCSca-1-injected hearts at 2 weeks post-intervention
show a significant improvement in left ventricle (LV) contractile capacity in iCPCSca-1-transplanted hearts. (C) Increased LV
wall thickness and decreased LV dilation in iCPCSca-1-injected animals, demonstrated by Masson’s Trichrome staining of LV
sections. Broken and continuous lines highlight the thickness of the infarcted LV wall in vehicle and iCPCSca-1 transplanted
animals, respectively. (D) Measurements of LV wall thickness in non-manipulated hearts and in the ischemic region corroborate
a significant difference between both experimental groups. Scale bar: 100 mm. Data are represented as mean – SEM
(***P < 0.001, **P < 0.01, and *P < 0.05 by Mann–Whitney test). Color images available online at www.liebertpub.com/scd
SCA-1
ICPC —A BENCHMARK FOR HEART SCA-11 PROGENITORS 9

attenuate LV remodeling after MI. However, the in vivo


differentiation of the CPCs in CM and endothelial cells is
minimal, and the amelioration observed on MI is primarily
attributed to paracrine effects [5]. Aiming at demonstrating
that iCPCSca-1 display the hallmarks of CPCs in the myo-
cardium in the onset of injury, a proof-of-principle ex-
periment was performed using a commonly reported
experimental injury setting [16,40]. Female C57BL/6 mice
subjected to MI via LAD coronary artery ligation were
immediately injected on the peri-infarct region with either
vehicle medium (n = 9) or male-derived iCPCSca-1 (n = 8).
MI size measurement (based on collagen deposition) at 14
days post-infarction showed that MI extent was compara-
ble between both experimental groups with 52% of the LV
wall being affected (Fig. 3A). Notwithstanding, echocar-
diography at 14 days, post-surgery in animals injected with
iCPCSca-1 showed improved LV function when compared
with vehicle controls, as demonstrated by significantly
increased (P < 0.05) EF and FS in iCPCSca-1-transplanted
hearts (Fig. 3B).
Altered cardiac loading after MI due to extensive loss
of myocardial cells induces changes in the heart shape,
size, and function that are commonly designated as cardiac
remodeling. A stereoscopic view of representative cross-
sections of infarcted hearts showed that iCPCSca-1 trans-
plantation prevented the major LV wall thinning and LV
chamber expansion observed in the vehicle control group
(Fig. 3C). This was corroborated by a morphometric anal-
ysis of MT stains, evidencing an increase (P < 0.01) in LV
wall thickness of iCPCSca-1-transplanted hearts (Fig. 3D) to
a value not statistically different from the non-manipulated FIG. 4. Contribution of iCPCSca-1 to neovascularization
animals (Fig. 3D). However and despite this increase, the post-MI. (A) Immunolocalization of CD31 in the ischemic
LV contractile function of iCPCSca-1-injected animals was region of control and iCPCSca-1-transplanted animals. Scale
not comparable with that of non-manipulated animals bar: 100 mm. (B) Quantification of CD31 + cells demon-
(P < 0.01, Fig. 3B). strates a significant increase in small vessels number in
Overall, the data suggest that iCPCSca-1 exerted a cardio- iCPCSca-1-injected animals. (C) Immunostaining combined
with fluorescence in situ hybridization (immuno-FISH)
protective effect, translated on a reduction of the deleterious
demonstrating iCPCSca-1 differentiation into CD31 + cells
consequences of ischemia and a partial recovery of cardiac (arrowheads). Scale bar: 10 mm. Data are represented as
functional parameters at 2 weeks post-MI. mean – SEM (***P < 0.001 by Mann–Whitney test). Color
images available online at www.liebertpub.com/scd
Transplanted iCPCSca-1 improve neovascularization
of the infarcted myocardium, engraft, and differentiate
into CM-, endothelial-, and smooth In fact, the majority of small vessels were not formed
muscle-like cells by donor cells, hinting that iCPCSca-1 contribution to neo-
vascularization is more likely to occur by stimulating the
To address whether cardiac performance amelioration mobilization and/or proliferation of host endothelial pre-
after iCPCSca-1 administration occurred at least partly via cursors/cells (paracrine action).
stimulation of the de novo vessel formation by paracrine We next investigated iCPCSca-1 functional engraftment
mechanisms, the density of small blood vessels in the in- and contribution to the de novo cardiac cells formation. In
farcted myocardium and in the border zone was quantified at vehicle-injected female mice, no Y-Chr + cells were de-
2 weeks post-surgery (Fig. 4A). Overall, iCPCSca-1 trans- tected in the heart (Fig. 5A); whereas in transplanted ani-
plantation resulted in a significant increase in CD31 + cells mals, numerous iCPCSca-1 were observed throughout
per mm2 when compared with the vehicle control (709 – 19 ischemic myocardium and MI border zone at 2 weeks post-
vs. 555 – 24, respectively, P < 0.001, Fig. 4B). iCPCSca-1, infarction. This demonstrates that cells were able to sur-
identified by Y-Chr staining were consistently observed in vive and engraft the LV ischemic region (Fig. 5B).
close proximity to vessels, suggesting contribution to either Moreover, iCPCSca-1 or their progeny were not found in
endothelial precursors mobilization/proliferation and/or healthy remote areas of the injury site. Ki67 staining was
differentiation into endothelial cells. The latter possibility frequently associated with Y-Chr evidencing iCPCSca-1
was evaluated by FISH combined with immunostaining for proliferation within the host myocardium (data not shown).
CD31, and, indeed, double positive events were observed in Assembly of the gap junction protein Cx43 at the cellular
all analyzed animals although at very low frequencies membrane confirmed that donor cells established func-
(4.1 – 0.7 cells per transverse heart section) (Figs. 4C and 5I). tional communication to both donor and host cells
10 FREIRE ET AL.

(Fig. 5C, D), substantiating successful iCPCSca-1 engraft- Co-localization of Y-Chr with a-SMA was observed in
ment within the host tissue. all analyzed animals (41.2 – 5.6 cells per transverse heart
Although the majority of grafted cells retained an undif- section) (Fig. 5I), either integrating small vessels (Fig. 5E)
ferentiated morphology and Sca-1 expression (Supplemen- or displaying a fibroblast-like morphology (Fig. 5F). In fact,
tary Fig. S4), discrete differentiation events were observed. in addition to smooth muscle cells, a-SMA is also expressed
by myofibroblasts, cells capable of establishing cell con-
nections through gap junctions, and relevant players in
formation of the contractile granulation tissue [41]. How-
ever, considering that Tcf21 is a marker of fibroblasts [42]
and is also involved in the activation of myofibroblasts [43],
and that iCPCSca-1 failed to express this marker even when
specifically stimulated (Figs. 1E and 2D), there is little
support for considering a myofibroblast fate. Furthermore,
iCPCSca-1 contributed to the cardiomyocytic compartment as
evidenced by a-actinin expression. The majority of a-actinin +
iCPCSca-1 presented an immature phenotype in three out of
six analyzed animals (9.0 – 0.6 cells per transverse heart
section) (Fig. 5I), suggesting an incomplete differentia-
tion. Moreover, a-actinin + iCPCSca-1 were found in close
proximity with a-actinin + host cells displaying a simi-
lar immature phenotype, suggesting an eventual iCPCSca-1—
mediated recruitment/activation of endogenous precursors
(Fig. 5G). Donor cells presenting well-defined cross-
striations, characteristic of a more mature phenotype,
were found in one animal (Fig. 5H; Supplementary Fig.
S5). While we do not rule out the possibility that these
scarce events resulted from cell fusion, differentiation
into more mature CM seems a more consistent scenario
(Supplementary Fig. S5).
Overall, these results are in agreement with the previous
observations, that Lin - Sca-1 + -mediated beneficial effects
after MI are partially mediated by paracrine mechanisms
[5,44] in detriment to a major contribution to cardiac cell
types after transplantation.

Discussion
Different progenitor cell subsets have been so far sug-
gested as candidates for the optimal cell population to be
implanted in infarcted and/or failing hearts (Fig. 6), but a
relationship among these populations remains undetermined.


FIG. 5. In vivo characterization of iCPCSca-1 on trans-
plantation into MI hearts. (A, B) Y-Chromosome detection
(FISH) in vehicle (A) and male-derived iCPCSca-1-injected
hearts (B) demonstrates the immortalized cell-line success-
ful engraftment. (C–H) Functional communication between
donor–donor [*, (C)] and donor–host [#, (D)] was revealed
by the assembly of Cx43 at the cellular membrane. iCPCSca-1
expressing a-SMA (arrowheads) were found integrating
small vessels [*, (E)] and scattered throughout the myo-
cardium displaying a fibroblast-like morphology (F). a-
Actinin + iCPCSca-1 (arrowheads) in the infarcted tissue
displayed an immature phenotype (G) and, less-frequently,
well-defined cross-striations (H). Scale bar: 10 mm. (I) Graph
displaying the number of cells, per transverse histological
section, bearing the Y-chr and expressing a-SMA, a-actinin,
or CD31. Data are represented as mean – SEM (***P < 0.001
by one-way ANOVA with post hoc Tukey’s test). Color
images available online at www.liebertpub.com/scd
SCA-1
ICPC —A BENCHMARK FOR HEART SCA-11 PROGENITORS 11

FIG. 6. Illustrative scheme of described multipotent progenitor cell populations isolated from the adult heart. Several
isolation methods have been applied to separate CPCs based on the expression of specific cell-surface markers (eg, c-Kit +
and Sca-1 + ), in vitro functional assays (eg, cardiospheres, side population), or the expression of a specific genetic marker
(Wt-1 epicardial progenitors). Regardless of the increasing number of putative CPCs reported, the functional, hierarchical,
and anatomical relationship of the illustrated cell progenitor subsets is still elusive. Color images available online at
www.liebertpub.com/scd

The absence of a specific molecular signature as well as the mental sets and the inter-laboratory comparison. Con-
multiplicity of the isolation and characterization protocols sistently, the present study has been carried out to create an
do not enable an understanding of whether the described immortalized CPC line retaining a constant phenotype cor-
CPC populations constitute different subsets of a common responding to native features. CPC immortalization has been
progenitor or independent cell lineages of distinct ontogeny. achieved by inducing TERT overexpression, in order to
Indeed, it has been suggested that the various identified circumvent the limitations determined by the diminished
CPCs partake in the same cell lineage, and the different mitotic capacity of somatic cells [48–52]. The use of im-
phenotypes are a consequence of the particular culturing mortalized lines that retain the in vivo features of primary
protocols used in different laboratories [21]. Importantly, cells, by combining the advantages of a high proliferative
other aspects account for an increased difficulty in the cell source with a stable phenotype in long-term culture, is a
identification of CPCs and to understand their real effi- major breakthrough in cell biology. Indeed, we have pre-
ciency. Namely, primary cells can easily change along time viously shown that bone marrow-derived mesenchymal
in culture as a consequence of genotypic and phenotypic stromal/stem cell (MSC) immortalized lines preserve mul-
drift and/or senescence [45–47] and, thus, lead to a lack of tipotency in standard and three-dimensional conditions in
reproducibility across studies. vitro [49,53]. Although telomerase overexpression does
Taken together, the experience accumulated in the last not induce oncogenesis, it may provide cells with a
decade indicates that only two strategies are used to im- greater opportunity to accumulate mutations, predisposing
plement a reliable cell therapy: to try and fail, that is, to to later malignant transformation [54,55]. The iCPCSca-1
implant whatever progenitor cell phenotype in the patient karyotype analysis showed aneuploidy, characteristic of
trusting to identify the perfect treatment, or to systematically malignant cells. However, an in vivo tumorigenicity assay
dig the complexity of the stem/progenitor cell behavior and, [30,31], monitored for 5 months, did not reveal tumor
based on novel data and concepts, to formulate a logically formation either locally (subcutaneously) or in any of the
stringent procedure to transfer experimental observations to analyzed organs (heart, spleen, lungs, and liver).
the clinical setting. Consistently with adult Lin - Sca-1 + CPC, the iCPCSca-1
Novel tools should be developed to help understanding phenotype was characterized by the expression of high
and, thus, governing the CPC complexity and phenotype levels of Sca-1, stemness-related genes (Bcrp1, Bmi-1 and
variability and to enable the standardization of the experi- Nestin), mesenchymal-associated proteins (eg, CD29,
12 FREIRE ET AL.

CD44, CD105, CD106, and PDGFRa), and early cardiac tion of new vessels in the infarcted tissue was carefully
transcription factors (Gata-4, Isl-1, Mef2c, and Tef-1). assessed. iCPCSca-1 transplanted hearts exhibited a denser
Conversely, immortalized cells did not express c-Kit while capillary network, when compared with the vehicle control
also lacking hematopoietic, endothelial, fibroblasts, mature group. In addition, iCPCSca-1 contributed directly to de novo
contractile myofilaments (a-and b-Mhc), and pluripotency vessel formation, as demonstrated by CD31 and a-SMA
(Sox2, Nanog, and Oct4) markers. expression on Y-Chr bearing cells. However, endothelial
iCPCSca-1 displayed the capability to generate in vitro differentiation was modest; hence, direct contribution of
cardiomyocyte-, smooth muscle-, and endothelial-like cells, engrafted cells to new vessel formation is minimal, sug-
with the differentiation toward smooth muscle-like cells gesting that paracrine mechanisms are involved in the re-
being the most efficient. This differential in vitro potential cruitment and/or proliferation of endothelial cells and/or
of Sca-1 + cells has been previously demonstrated by others precursors.
[16]. Whether this indicates a higher commitment of CPCs differentiation into CM has been frequently dem-
CPCSca-1 to smooth muscle-like cells or to the fact that the onstrated by the expression of proteins of the contractile
in vitro differentiation conditions are less favorable to ac- machinery [5,6,12,15]. Indeed, although CPCs appear to
tivate endothelial and cardiomyocytic pathways, and, thus, grasp a lot of expectations with regard to cardiac cell ther-
not suitable to demonstrate the full potential of these cells, apies, most studies still report an immature phenotype for
still needs to be determined. the differentiated cells, to a great extent resembling fetal-
iCPCSca-1 is a phenotypically stable cell line that provides neonatal CM [5,6,12,15]. In our in vivo setting, iCPCSca-1
investigators with the same unique cell model to dissect differentiated into cardiomyocyte-like cells, though the
CPC behavior and differentiation mechanisms. Furthermore, majority displayed a disorganized structure with no detect-
iCPCSca-1 could represent a benchmark to compare with able sarcomeres, characteristic of an immature phenotype. It
other cells posited to bear stemness characteristics. In fact, should be noted that both primary CPCSca-1 and iCPCSca-1
the immortalized CPCs retain the features of their ex vivo express the transcription factor c-Myc, which has been
counterparts, that is, moderate differentiation into cardio- previously shown to play a role in the blockage of cell
myocyte-, endothelial-, and smooth muscle-like cells, when differentiation [36] and in the survival and proliferation of
subjected to appropriate stimuli in vitro as well as in vivo, in CPCs [37]. Whether the levels of c-Myc are regulating
a myocardial injury setting. Furthermore, the cell line CPCs cell-fate decision to differentiate at the expenses of
combines the advantages of a high proliferative cell source cell proliferation remains to be determined and should be
with a stable phenotype in long-term culture. Indeed, murine subject to further investigation. Since iCPCSca-1 showed an
cells are prone to undergo spontaneous immortalization after in vitro and in vivo differentiation potential similar to their
prolonged in vitro culture [46,56,57], which may result native counterparts, it is our conviction that iCPCSca-1
in the accumulation of unpredictable cytogenetic modifica- constitutes a reliable easy-to-use model to dissect the key
tions and malignant transformations [47]. The use of un- molecular mechanisms governing cardiomyocytic differen-
characterized spontaneously immortalized cell clones as in tiation from adult CPCs.
vitro models for their native cell counterparts should be In this work, an immortalized cell line for modeling
circumvented due to the dissimilar phenotypes generated by mouse Lin - Sca-1 + CPCs was specifically generated by
unpredictable cytogenetic alterations in overextended sub- means of TERT overexpression. The immortalization did
culture in vitro. not appear to impair any important cellular property,
The iCPCSca-1 capability to mimic their native non- as iCPCSca-1 were phenotypically and functionally similar to
immortalized corresponding cells has also been apparent in their primary cell counterparts. Overall, the results indicate
in vivo experiments. In fact, iCPCSca-1 were able to con- that iCPCSca-1 are representative of the native Sca-1 + car-
tribute toward repairing the myocardial injuries experi- diac population, thus constituting a suitable tool for func-
mentally provoked by coronary ligation in vivo, as shown by tional and mechanistic studies in need in the field. Hence,
minor LV chamber expansion and increased thickness of the with regard to the heterogeneity and low frequency in the
LV-free wall. Importantly, echocardiography follow-up re- adult heart of described CPC populations (Fig. 6), iCPCSca-1
vealed improved systolic function after iCPCSca-1 trans- comes forth as a uniquely validated off-the-shelf line for in
plantation into MI animals. iCPCSca-1 abundance in the host vitro high-throughput and bioengineering studies. The latter
infarcted and peri-infarcted myocardium was particularly combined with the remarkable capacity of iCPCSca-1 to en-
surprising, and cells were able not only to survive but also to graft and differentiate in loco makes this cell line a valuable
engraft. Moreover, engrafted cells established donor–donor model system to investigate in vivo the role of the Sca-1 +
and donor–host Cx43-mediated connections. Indeed, Cx43 stem/progenitor cells resident in the adult heart.
promotes MSC survival in the ischemic heart [58], and it is Last but not the least, the widespread use of this and other
critical for protection against ventricular tachycardia, which similarly generated cardiac progenitor-subset cell lines will
is a resultant from the transplantation of embryonic CM in enable the reduction of animal usage and will contribute in a
MI [59]. Accordingly, it can be conjectured that Cx43 is properly standardized manner to the definition of bench-
involved in iCPCSca-1 resistance to the adverse scenario of mark(s) for stem/progenitor cells from distinct organ sys-
myocardial ischemia. tems and from different laboratories.
The implantation of immortalized cells into infarcted
mouse hearts also induced a significant improvement in the
Acknowledgments
vascularization of the damaged region, very likely, through
a paracrine action. Since an improved vascularization is This work was supported by Fundação para a Ciência e a
associated with a ‘‘pro-regenerative’’ response, the forma- Tecnologia (SFRH/BD/64715/2009 to A.G.F., SFRH/BPD/
SCA-1
ICPC —A BENCHMARK FOR HEART SCA-11 PROGENITORS 13

42254/2007 to D.S.N., SFRH/BD/74218/2010 to M.V., myocardial stem cell population. FEBS Lett 530:239–
SFRH/BPD/80588/2011 to T.P.R., and Ciência2007 to 243.
P.P.O.); Fundo Europeu de Desenvolvimento Regional, 10. Martin CM, AP Meeson, SM Robertson, TJ Hawke,
Programa Operacional Factores de Competitividade- JA Richardson, S Bates, SC Goetsch, TD Gallardo and
COMPETE, Quadro de Referência Estratégico Nacional, DJ Garry. (2004). Persistent expression of the ATP-
and Fundo Social Europeu (PEst-C/SAU/LA0002/2011, binding cassette transporter, Abcg2, identifies cardiac SP
PTDC/SAU-OSM-68473/2006, PTDC/SAU-ORG/118297/ cells in the developing and adult heart. Dev Biol 265:
2010 and NORTE-07-0124-FEDER-000005 to D.S.N.); and 262–275.
European Regional Development Fund – Project FNUSA- 11. Messina E, L De Angelis, G Frati, S Morrone, S Chimenti,
ICRC (no. CZ.1.05/1.1.00/02.0123). Travel funds were F Fiordaliso, M Salio, M Battaglia, MV Latronico, et al.
(2004). Isolation and expansion of adult cardiac stem cells
awarded by Boehringer Ingelheim Fonds to (T.P.R.); by
from human and murine heart. Circ Res 95:911–921.
American Portuguese Biomedical Research Fund to (P.P.O
12. Oh H, SB Bradfute, TD Gallardo, T. Nakamura, V. Gaus-
and D.S.N.). sin, Y Mishina, J Pocius, LH Michael, RR Behringer, et al.
The authors are indebted to Joana G. Guedes (CIBIO), (2003). Cardiac progenitor cells from adult myocardium:
Rui Fernandes (IBMC), Paula Sampaio (IBMC), Dina Lei- homing, differentiation, and fusion after infarction. Proc
tão (IPATIMUP), and the members of MCA Lab (IBMC) Natl Acad Sci U S A 100:12313–12318.
for sharing scientific expertise and equipment. 13. Pfister O, F Mouquet, M Jain, R Summer, M Helmes, A
Fine, WS Colucci and R Liao. (2005). CD31- but not
CD31 + cardiac side population cells exhibit functional
Author Disclosure Statement cardiomyogenic differentiation. Circ Res 97:52–61.
No competing financial interests exist. 14. Smart N, S Bollini, KN Dube, JM Vieira, B Zhou, S Da-
vidson, D Yellon, J Riegler, AN Price, et al. (2011). De
novo cardiomyocytes from within the activated adult heart
References after injury. Nature 474:640–644.
15. Takamiya M, KH Haider and M Ashraf. (2011). Iden-
1. Bui QT, ZM Gertz and RL Wilensky. (2010). Intracoronary tification and characterization of a novel multipotent
delivery of bone-marrow-derived stem cells. Stem Cell Res sub-population of Sca-1 + cardiac progenitor cells for
Ther 1:29. myocardial regeneration. PLoS One 6:e25265.
2. Muller-Ehmsen J, B Krausgrill, V Burst, K Schenk, UC 16. Tateishi K, E Ashihara, N Takehara, T Nomura, S Hon-
Neisen, JW Fries, BK Fleischmann, J. Hescheler and RH sho, T Nakagami, S Morikawa, T Takahashi, T Ueyama,
Schwinger. (2006). Effective engraftment but poor mid- H Matsubara and H Oh. (2007). Clonally amplified
term persistence of mononuclear and mesenchymal bone cardiac stem cells are regulated by Sca-1 signaling for
marrow cells in acute and chronic rat myocardial infarction. efficient cardiovascular regeneration. J Cell Sci 120:
J Mol Cell Cardiol 41:876–884. 1791–1800.
3. Gnecchi M, H He, OD Liang, LG Melo, F Morello, H Mu, 17. Yamahara K, S Fukushima, SR Coppen, LE Felkin,
N Noiseux, L Zhang, RE Pratt, JS Ingwall and VJ Dzau. A Varela-Carver, PJR Barton, MH Yacoub and K Suzuki.
(2005). Paracrine action accounts for marked protection of (2008). Heterogeneic nature of adult cardiac side popu-
ischemic heart by Akt-modified mesenchymal stem cells. lation cells. Biochem Biophys Res Commun 371:615–
Nat Med 11:367–368. 620.
4. Kinnaird T, E Stabile, MS Burnett, M Shou, CW Lee, S 18. Ye J, A Boyle, H Shih, RE Sievers, Y Zhang, M Prasad,
Barr, S Fuchs and SE Epstein. (2004). Local delivery of H Su, Y Zhou, W Grossman, HS Bernstein and
marrow-derived stromal cells augments collateral perfusion Y Yeghiazarians. (2012). Sca-1 + cardiosphere-derived
through paracrine mechanisms. Circulation 109:1543– cells are enriched for Isl1-expressing cardiac precursors and
1549. improve cardiac function after myocardial injury. PLoS
5. Wang X, Q Hu, Y Nakamura, J Lee, G Zhang, AH From One 7:e30329.
and J Zhang. (2006). The role of the sca-1 + /CD31 - cardiac 19. Bolli R, AR Chugh, D D’Amario, JH Loughran, MF
progenitor cell population in postinfarction left ventricular Stoddard, S Ikram, GM Beache, SG Wagner, A Leri, et al.
remodeling. Stem Cells 24:1779–1788. (2011). Cardiac stem cells in patients with ischaemic car-
6. Beltrami AP, L Barlucchi, D Torella, M Baker, F Limana, diomyopathy (SCIPIO): initial results of a randomised
S Chimenti, H Kasahara, M Rota, E Musso, et al. (2003). phase 1 trial. Lancet 378:1847–1857.
Adult cardiac stem cells are multipotent and support 20. Lim GB. (2012). Stem cells: myocardial regeneration after
myocardial regeneration. Cell 114:763–776. infarction—promising phase I trial results. Nat Rev Cardiol
7. Chong JJ, V Chandrakanthan, M Xaymardan, NS Asli, J Li, 9:187.
I Ahmed, C Heffernan, MK Menon, CJ Scarlett, et al. 21. Di Nardo P, G Forte, A Ahluwalia and M Minieri. (2010).
(2011). Adult cardiac-resident msc-like stem cells with a Cardiac progenitor cells: potency and control. J Cell Phy-
proepicardial origin. Cell Stem Cell 9:527–540. siol 224:590–600.
8. Gambini E, G Pompilio, A Biondi, F Alamanni, MC 22. Di Nardo P and GC Parker. (2011). Stem cell standardi-
Capogrossi, M Agrifoglio and M Pesce. (2011). C-kit + zation. Stem Cells Dev 20:375–377.
cardiac progenitors exhibit mesenchymal markers and 23. Dawn B and R Bolli. (2005). Cardiac progenitor cells. Circ
preferential cardiovascular commitment. Cardiovasc Res Res 97:1080–1082.
89:362–373. 24. Gonzalez A, M Rota, D Nurzynska, Y Misao, J Tillmanns,
9. Hierlihy AM, P Seale, CG Lobe, MA Rudnicki and C Ojaimi, ME Padin-Iruegas, P Müller, G Esposito, et al.
LA Megeney. (2002). The post-natal heart contains a (2008). Activation of cardiac progenitor cells reverses the
14 FREIRE ET AL.

failing heart senescent phenotype and prolongs lifespan. marrow-derived stem cells with cardiomyocytes in a het-
Circ Res 102:597–606. erologous in vitro model. Eur J Cardio-Thorac Surg
25. Hooijberg E, JJ Ruizendaal, PJF Snijders, EWM Kueter, 28:685–691.
JMM Walboomers and H Spits. (2000). Immortalization 39. Wurmser AE and FH Gage. (2002). Stem cells: cell fusion
of human CD8 + T cell clones by ectopic expression of causes confusion. Nature 416:485–487.
telomerase reverse transcriptase. J Immunol 165:4239– 40. Liang SX, TYL Tan, L Gaudry and B Chong. (2010).
4245. Differentiation and migration of Sca1 + /CD31 - cardiac
26. Simonsen JL, C Rosada, N Serakinci, J Justesen, K Sten- side population cells in a murine myocardial ischemic
derup, SIS Rattan, TG Jensen and M Kassem. (2002). model. Int J Cardiol 138:40–49.
Telomerase expression extends the proliferative life-span 41. Baum J and HS Duffy. (2011). Fibroblasts and myofibro-
and maintains the osteogenic potential of human bone blasts: what are we talking about? J Cardiovasc Pharmacol
marrow stromal cells. Nat Biotechnol 20:592–596. 57:376–379.
27. Yang J, E Chang, AM Cherry, CD Bangs, Y Oei, A Bodnar, 42. Lajiness JD and SJ Conway. (2013). Origin, development,
A Bronstein, C-P Chiu and GS Herron. (1999). Human and differentiation of cardiac fibroblasts. J Mol Cell Cardiol
endothelial cell life extension by telomerase expression. J pii:S0022–2828.
Biol Chem 274:26141–26148. 43. Plotkin M and V Mudunuri. (2008). Pod1 induces myofi-
28. Jiang X-R, G Jimenez, E Chang, M Frolkis, B Kusler, M broblast differentiation in mesenchymal progenitor cells
Sage, M Beeche, AG Bodnar, GM Wahl, TD Tlsty and C-P from mouse kidney. J Cell Biochem 103:675–690.
Chiu. (1999). Telomerase expression in human somatic 44. Matsuura K, A Honda, T Nagai, N Fukushima, K Iwanaga,
cells does not induce changes associated with a transformed M Tokunaga, T Shimizu, T Okano, H Kasanuki, N Hagi-
phenotype. Nat Genet 21:111–114. wara and I Komuro. (2009). Transplantation of cardiac
29. Forte G, F Carotenuto, F Pagliari, S Pagliari, P Cossa, R progenitor cells ameliorates cardiac dysfunction after
Fiaccavento, A Ahluwalia, G Vozzi, B Vinci, et al. (2008). myocardial infarction in mice. J Clin Invest 119:2204–
Criticality of the biological and physical stimuli array in- 2217.
ducing resident cardiac stem cell determination. Stem Cells 45. Macieira-Coelho A, E Loria and L Berumen. (1975). Re-
26:2093–2103. lationship between cell kinetic changes and metabolic
30. Schou M and A-M Engel. (2006). Assay of tumorigenicity events during cell senescence in vitro. Adv Exp Med Biol
in nude mice. In: Cell Biology: a laboratory handbook. JE 53:51–65.
Cells, ed. Elsevier Science, pp 353–357. 46. Sherr CJ and RA DePinho. (2000). Cellular senescence:
31. Wu G-J, P Fu, S-W Wang and M-WH Wu. (2008). En- minireview mitotic clock or culture shock? Cell 102:407–
forced expression of MCAM/MUC18 Increases in vitro 410.
motility and invasiveness and in vivo metastasis of two 47. Tolar J, AJ Nauta, MJ Osborn, A Panoskaltsis Mortari, RT
mouse melanoma K1735 sublines in a syngeneic mouse McElmurry, S Bell, L Xia, N Zhou, M Riddle, et al. (2007).
model. Mol Cancer Res 6:1666–1677. Sarcoma derived from cultured mesenchymal stem cells.
32. Smits AM, P van Vliet, CH Metz, T Korfage, JPG Sluijter, Stem Cells 25:371–379.
PA Doevendans and M-J Goumans. (2009). Human cardi- 48. Armstrong L, G Saretzki, H Peters, I Wappler, J Evans, N
omyocyte progenitor cells differentiate into functional Hole, T von Zglinicki and M Lako. (2005). Overexpression
mature cardiomyocytes: an in vitro model for studying of telomerase confers growth advantage, stress resistance,
human cardiac physiology and pathophysiology. Nat Protoc and enhanced differentiation of ESCs toward the hemato-
4:232–243. poietic lineage. Stem Cells 23:516–529.
33. Pagliari S, AC Vilela-Silva, G Forte, F Pagliari, C Man- 49. Forte G, O Franzese, S Pagliari, F Pagliari, AM Di Fran-
doli, G Vozzi, S Pietronave, M Prat, S Licoccia, et al. cesco, P Cossa, A Laudisi, R Fiaccavento, M Minieri, E
(2011). Cooperation of biological and mechanical signals Bonmassar and P Di Nardo. (2009). Interfacing Sca-1(pos)
in cardiac progenitor cell differentiation. Adv Mater mesenchymal stem cells with biocompatible scaffolds with
23:514–518. different chemical composition and geometry. J Biomed
34. Michael LH, ML Entman, CJ Hartley, KA Youker, J Zhu, Biotechnol 2009:910610.
SR Hall, HK Hawkins, K Berens and CM Ballantyne. 50. Geserick C, A Tejera, E Gonzalez-Suarez, P Klatt and MA
(1995). Myocardial ischemia and reperfusion: a murine Blasco. (2006). Expression of mTert in primary murine
model. Am J Physiol 269:H2147–H2154. cells links the growth-promoting effects of telomerase to
35. Nascimento DS, M Valente, T Esteves, F de Pina Mde, transforming growth factor-beta signaling. Oncogene 25:
JG Guedes, A Freire, P Quelhas and P Pinto-do-Ó. 4310–4319.
(2011). MIQuant: semi-automation of infarct size as- 51. Lee MK, MP Hande and K Sabapathy. (2005). Ectopic
sessment in models of cardiac ischemic injury. PLoS mTERT expression in mouse embryonic stem cells does
One 6:e25045. not affect differentiation but confers resistance to differ-
36. Bhattacharyya S, A Kumar and K Lal Khanduja. (2012). entiation- and stress-induced p53-dependent apoptosis. J
The voyage of stem cell toward terminal differentiation: a Cell Sci 118:819–829.
brief overview. Acta Biochim Biophys Sin (Shanghai) 52. Steele SL, Y Wu, RJ Kolb, M Gooz, CJ Haycraft, KT
44:463–475. Keyser, L Guay-Woodford, H Yao and PD Bell. (2010).
37. Cottage CT, B Bailey, KM Fischer, D Avitable, B Collins, Telomerase immortalization of principal cells from mouse
S Tuck, P Quijada, N Gude, R Alvarez, J Muraski and MA collecting duct. Am J Physiol Renal Physiol 299:F1507–
Sussman. (2010). Cardiac progenitor cell cycling stimu- F1514.
lated by Pim-1 kinase. Circ Res 106:891–901. 53. Miranda CO, CA Teixeira, MA Liz, VF Sousa, F Fran-
38. Garbade J, A Schubert, AJ Rastan, D Lenz, T Walther, JF quinho, G Forte, P Di Nardo, OP Pinto-Do and MM Sousa.
Gummert, S Dhein and F-W Mohr. (2005). Fusion of bone (2011). Systemic delivery of bone marrow-derived mes-
SCA-1
ICPC —A BENCHMARK FOR HEART SCA-11 PROGENITORS 15

enchymal stromal cells diminishes neuropathology in a Address correspondence to:


mouse model of Krabbe’s disease. Stem Cells 29:1738– Dr. Perpétua Pinto-do-Ó
1751. INEB—Instituto de Engenharia Biomédica
54. Harley CB. (2002). Telomerase is not an oncogene. On- Universidade do Porto
cogene 21:494–502. Rua do Campo Alegre, 823
55. Prescott JC and EH Blackburn. (1999). Telomerase: Dr. Porto 4150-180
Jekyll or Mr. Hyde? Curr Opin Genet Dev 9:368–373. Portugal
56. Denhardt DT, DR Edwards, M McLeod, G Norton, CLJ
Parfett and M Zimmer. (1991). Spontaneous immortaliza- E-mail: perpetua@ineb.up.pt
tion of mouse embryo cells: strain differences and changes
in gene expression with particular reference to retroviral
gag-pol genes. Exp Cell Res 192:128–136. Paolo Di Nardo
57. Fusenig NE, RT Dzarlieva-Petrusevska and D Breitkreutz. Laboratorio di Cardiologia Molecolare and Cellulare
(1985). Phenotypic and cytogenetic characteristics of
Dip. di Medicina Interna
different stages during spontaneous transformation of
Università di Roma Tor Vergata
mouse keratinocytes in vitro. Carcinog Compr Surv 9:
293–326.
Via Montpellier
58. Wang D, W Shen, F Zhang, M Chen, H Chen and K Roma 100133
Cao. (2010). Connexin43 promotes survival of mesen- Italy
chymal stem cells in ischaemic heart. Cell Biol Int 34:
415–423. E-mail: dinardo@med.uniroma2.it
59. Roell W, T Lewalter, P Sasse, YN Tallini, B-R Choi, M
Breitbach, R Doran, UM Becher, S-M Hwang, et al. (2007). Received for publication July 16, 2013
Engraftment of connexin 43-expressing cells prevents post- Accepted after revision December 21, 2013
infarct arrhythmia. Nature 450:819–824. Prepublished on Liebert Instant Online XXXX XX, XXXX

También podría gustarte