You are on page 1of 14

Integrative Physiology

Induced Pluripotent Stem Cell (iPSC)–Derived


Extracellular Vesicles Are Safer and More Effective
for Cardiac Repair Than iPSCs
Marta Adamiak,* Guangming Cheng,* Sylwia Bobis-Wozowicz, Lin Zhao,
Sylwia Kedracka-Krok, Anweshan Samanta, Elzbieta Karnas, Yu-Ting Xuan,
Bozena Skupien-Rabian, Xing Chen, Urszula Jankowska, Magdy Girgis, Malgorzata Sekula,
Arash Davani, Slawomir Lasota, Robert J. Vincent, Michal Sarna, Kathy L. Newell,
Ou-Li Wang, Nathaniel Dudley, Zbigniew Madeja, Buddhadeb Dawn, Ewa K. Zuba-Surma

Rationale: Extracellular vesicles (EVs) are tiny membrane-enclosed droplets released by cells through membrane
budding or exocytosis. The myocardial reparative abilities of EVs derived from induced pluripotent stem cells
(iPSCs) have not been directly compared with the source iPSCs.
Objective: To examine whether iPSC-derived EVs can influence the biological functions of cardiac cells in vitro and
to compare the safety and efficacy of iPSC-derived EVs (iPSC-EVs) and iPSCs for cardiac repair in vivo.
Methods and Results: Murine iPSCs were generated, and EVs isolated from culture supernatants by sequential
centrifugation. Atomic force microscopy, high-resolution flow cytometry, real-time quantitative RT-PCR, and
mass spectrometry were used to characterize EV morphology and contents. iPSC-EVs were enriched in miRNAs
and proteins with proangiogenic and cytoprotective properties. iPSC-EVs enhanced angiogenic, migratory, and
antiapoptotic properties of murine cardiac endothelial cells in vitro. To compare the cardiac reparative capacities
in vivo, vehicle, iPSCs, and iPSC-EVs were injected intramyocardially at 48 hours after a reperfused myocardial
infarction in mice. Compared with vehicle-injected mice, both iPSC- and iPSC-EV–treated mice exhibited improved
left ventricular function at 35 d after myocardial infarction, albeit iPSC-EVs rendered greater improvement.
iPSC-EV injection also resulted in reduction in left ventricular mass and superior perfusion in the infarct zone.
Downloaded from http://ahajournals.org by on December 18, 2020

Both iPSCs and iPSC-EVs preserved viable myocardium in the infarct zone, whereas reduction in apoptosis was
significant with iPSC-EVs. iPSC injection resulted in teratoma formation, whereas iPSC-EV injection was safe.
Conclusions: iPSC-derived EVs impart cytoprotective properties to cardiac cells in vitro and induce superior cardiac
repair in vivo with regard to left ventricular function, vascularization, and amelioration of apoptosis and hypertrophy.
Because of their acellular nature, iPSC-EVs represent a safer alternative for potential therapeutic applications in
patients with ischemic myocardial damage.   (Circ Res. 2018;122:296-309. DOI: 10.1161/CIRCRESAHA.117.311769.)
Key Words: angiogenesis ■ apoptosis ■ extracellular vesicles ■ induced pluripotent stem cells
■ myocardial infarction ■ remodeling ■ stem cells

B ecause the adult mammalian heart possesses limited regen-


erative capacity after injury, cell therapy has emerged as
a potential approach for repair of the infarcted heart. Several
Editorial, see p 197
In This Issue, see p 185
Meet the First Author, see p 186
adult stem and progenitor cells from various sources have al-
ready been tested in patients, albeit with modest benefits.1 that in addition to cellular replacement, paracrine factors re-
Although induced pluripotent stem cells (iPSCs) offer exciting leased by injected cells are also important for cell-based heart
opportunities for tissue restoration, recent evidence indicates repair.2 These paracrine factors may be secreted directly from

Original received July 21, 2017; revision received November 3, 2017; accepted November 7, 2017. In October 2017, the average time from submission
to first decision for all original research papers submitted to Circulation Research was 13 days.
From the Department of Cell Biology (M.A., S.B.-W., E.K., S.L., Z.M., E.K.Z.-S), Department of Physical Biochemistry (S.K.-K., B.S.-R.), and
Department of Biophysics (M. Sarna), Jagiellonian University, Krakow, Poland; Division of Cardiovascular Diseases, Cardiovascular Research Institute
(G.C., L.Z., A.S., Y.-T.X., X.C., M.G., A.D., R.J.V., O.-L.W., N.D., B.D.) and Department of Pathology and Laboratory Medicine (K.L.N.), University of
Kansas Medical Center, Kansas City; and Malopolska Centre of Biotechnology, Krakow, Poland (E.K., B.S.-R., U.J., M. Sekula, M. Sarna).
*These authors contributed equally to this article.
The online-only Data Supplement is available with this article at http://circres.ahajournals.org/lookup/suppl/doi:10.1161/CIRCRESAHA.
117.311769/-/DC1.
Correspondence to Ewa K. Zuba-Surma, PhD, DSc, Prof JU, Department of Cell Biology, Jagiellonian University, Krakow 30-663, Poland, E-mail ewa.
zuba-surma@uj.edu.pl; or Buddhadeb Dawn, MD, Division of Cardiovascular Diseases, Cardiovascular Research Institute, University of Kansas Medical
Center, Kansas City, KS 66160, E-mail bdawn@kumc.edu
© 2017 American Heart Association, Inc.
Circulation Research is available at http://circres.ahajournals.org DOI: 10.1161/CIRCRESAHA.117.311769

296
Adamiak et al   iPSC-EVs for Myocardial Repair   297

Novelty and Significance


What Is Known? iPSCs, which are able to give rise to numerous cell types, have
shown great promise for heart regeneration. EVs, minute mem-
• Induced pluripotent stem cells (iPSCs) differentiate into numerous cell
types and, therefore, hold great promise for heart regeneration.
brane-enclosed droplets released by mammalian cells, have also
• Extracellular vesicles (EVs), including the fraction of exosomes derived been shown to ameliorate myocardial ischemia/reperfusion in-
from various stem cells, have been shown to ameliorate myocardial jury. However, the myocardial reparative abilities of EVs derived
ischemia/reperfusion injury and promote cardiac repair. from iPSCs have not been directly compared with the source
• The comparative safety and efficacy of iPSCs and iPSC-derived EVs iPSCs. The results from quantitative RT-PCR and proteomic analy-
(iPSC-EVs) for infarct repair have not been examined. ses revealed that EV cargo is enriched with numerous molecules
that impart cytoprotective and angiogenic benefits to cardiac
What New Information Does This Article Contribute? cells. After ischemia/reperfusion and transplantation in vivo, both
iPSC- and iPSC-EV–treated mice exhibited improved LV function
• The molecular cargo within iPSC-EVs includes numerous miRNAs that
compared with vehicle-treated mice; however, iPSC-EVs induced
are present in iPSCs and several additional miRNAs that are enriched
greater functional benefits and resulted in superior perfusion in
specifically in EVs. These miRNAs are known to promote cellular
the infarct zone and prevented apoptosis. iPSC injections resulted
proliferation and differentiation, enhance angiogenesis, and prevent
apoptosis.
in teratoma formation, whereas iPSC-EV injections were safe.
• The proteomic analysis of EVs revealed molecules that stimulate car- These observations indicate that iPSC-EVs confer cytoprotective
diomyogenesis; promote cardiac, endothelial, and smooth muscle cell properties to cardiac cells in vitro and induce superior cardiac
proliferation; and protect against oxidative damage. repair in vivo without the risk of teratoma formation seen with
• The iPSC-EVs induced angiogenic, migratory, and antiapoptotic prop- parental iPSCs. Thus, iPSC-EVs offer a safer alternative for poten-
erties in cardiac endothelial cells in vitro and induced superior infarct tial therapeutic applications in patients with ischemic myocardial
repair in vivo compared with iPSCs. iPSC injection resulted in teratoma damage.
formation, whereas EV injection did not produce any tumor.

compared head-to-head, for the first time, the safety and effi-
Nonstandard Abbreviations and Acronyms
cacy of iPSC-EVs with their parental iPSCs for cardiac repair
EVs extracellular vesicles in vivo. Our results show that iPSC-EVs carry unique sets of
proteins and regulatory miRNAs and impact by angiogenic
Downloaded from http://ahajournals.org by on December 18, 2020

iPSC induced pluripotent stem cell


iPSC-EV induced pluripotent stem cell–derived extracellular vesicles capacity, migratory activity, and survival of cardiac endothe-
LV left ventricle lial cells (CECs) in cytotoxic environment. In vivo, injection
MI myocardial infarction of iPSC-EVs after acute myocardial infarction (MI) improves
left ventricular (LV) function and remodeling. Importantly, we
provide evidence that although injection of iPSCs leads to tu-
the transplanted cells or released as cargo in tiny membrane-en- mor formation, the use of EVs derived from these cells is safe.
closed structures termed extracellular vesicles (EVs).2,3 Given
the potential of both iPSCs and iPSC-derived EVs (iPSC-EVs) Methods
for myocardial repair, there is a strong rationale to directly com- An expanded methods section is available in the Online Data
pare the safety and reparative efficacy of these 2 approaches. Supplement.
The authors declare that all data that support the findings of this
EVs represent small (30–1000 nm) vesicles released by study are available within the article and its Online Data Supplement.
cells either by outward budding from the plasma membrane or The present study was performed in accordance with the guide-
via exocytosis.4 Upon shedding, EVs may fuse with the recipi- lines of the Animal Care and Use Committee of the University of
ent cells directly or after endocytosis followed by the release of Kansas Medical Center and with the Guide for the Care and Use of
contents or interact with target cells by receptor–ligand inter- Laboratory Animals (Department of Health and Human Services, pub-
lication No. [National Institutes of Health] 86-23) and also in accor-
actions.5,6 Because EVs carry proteins, mRNAs, and miRNAs, dance with the Polish and European legislation after approval by the
from their parental cells, they represent an important compo- First Local Ethical Committee on Animal Testing at the Jagiellonian
nent of paracrine signaling and cell-to-cell communication.7 University. The data that support the findings of this study are avail-
Although the regenerative abilities of EVs harvested from mes- able from the corresponding authors on reasonable request.
enchymal stem cells and endothelial and cardiac progenitors
have been reported,8–10 the biological basis of EV actions needs iPSC Generation
Murine fibroblasts were transfected with lentivirus expressing Oct4,
further elucidation. Thus, another major rationale for this study Sox2 (sex-determining region Y-box 2), and Klf4. iPSC colonies were
was to characterize the molecular contents of EVs with regard picked, further expanded, and adapted to serum-free conditions. Colonies
to potential paracrine activities relevant for myocardial repair. with high contents of pluripotency-associated transcripts (Online Figure
Finally, although iPSCs have been known to produce terato- I) were chosen for further studies. iPSCs were phenotyped by flow cy-
mas,11 the safety of iPSC-EVs remains unknown. tometry and immunocytochemistry following standard protocols.
Accordingly, we meticulously characterized the morphol- EV Purification
ogy and contents of iPSC-EVs with a multi-instrumental EVs were prepared from the culture supernatant of iPSCs using a pro-
approach, examined their biological function in vitro, and tocol optimized in our laboratory (Online Figure II).12 Nanoparticle
298  Circulation Research  January 19, 2018

tracking analysis, atomic force microscopy, high-resolution flow cytome- pluripotency-related markers, including Oct3/4A, Nanog,
try, and quantitative RT-PCR were used to characterize EVs. MicroRNA Sox2, and Rex1, at the mRNA and protein levels, as well as
expression profiling was performed using quantitative RT-PCR.
placental alkaline phosphatase (Figure 1A through 1C; Online
EV Protein Analysis by LC-MS/MS Figure I). iPSCs also expressed adhesion molecules and
After lysis, samples from iPSCs and iPSC-EVs for LC-MS/MS (liquid growth factor receptors considered important for stem cell ac-
chromatography–mass spectrometry) analysis were prepared using tivities, including CD49e, CD29, CD105, and CD117, which
the FASP (filter-aided sample preparation) method.13 Peptide fractions may be potentially transferred to iPSC-EVs (Figure 1D).
were analyzed using Q Exactive high-resolution mass spectrometer
These results provide evidence that pluripotent iPSCs can be
coupled with nanoHPLC. The data were analyzed using Proteome
Discoverer 1.4 and a MASCOT server against the Swissprot_201509 effectively cultured in feeder- and serum-free conditions for
database. Search result validation was performed using Percolator al- uncontaminated harvest of EVs.
gorithm.14 Gene ontology (GO) was analyzed using FatiGO software. Next, iPSC-EVs were successfully isolated from the iPSC-
conditioned media by a sequential centrifugation procedure,
Assessment of EV Function In Vitro including ultracentrifugation, resulting in EV specimens con-
Murine CECs and fluorescent EVs were used for cellular uptake stud-
ies. Capillary tube formation assay was performed on Matrigel. The taining both microvesicles and exosome fractions (Figure 2A).
movement of CECs was time-lapse recorded, and cell trajectories Importantly, we confirmed the presence of iPSC-EVs accord-
were constructed and analyzed. CEC viability and apoptosis were ing to the current recommendations18 by examining their (1)
analyzed by flow cytometry. size distribution by nanoparticle tracking analysis platform, (2)
high-resolution morphology and membrane elasticity by atom-
Experimental Protocol for the In Vivo Studies
A total of 43 wild-type (C57BL6/J) mice were used for the in vivo ic force microscopy, and (3) expression of EV-related antigens
studies. The experimental protocol is summarized in Online Figure (eg, CD9 and CD81 tetraspanins) by Western and high-reso-
III. All mice underwent a 30-minute coronary occlusion followed by lution flow cytometry (Figure 2B through 2E). Moreover, we
reperfusion. After 48 hours, chest was reopened, and mice received confirmed the iPSC origin by detecting transcripts for pluripo-
intramyocardial injection of vehicle (group I), iPSCs (group II), or
tent transcription factors Oct3/4, Nanog, and Rex1 (Figure 2F).
iPSC-EVs (group III). Echocardiographic studies were performed 4
days before coronary occlusion/reperfusion, at 48 hours after cell in- These data confirm that the iPSC-EV specimens used in further
jection, and 35 days after MI using a Vevo 2100 Ultrasound System.15 studies represent homogenous vesicular fractions that exhibit
common EV morphology and antigen expression, along with
Morphometry and Immunohistochemistry the presence of iPSC-derived markers.
After 35 days, mice were euthanized and hearts harvested and for-
malin fixed. All morphometric and immunohistochemical analyses iPSC-EVs Contained iPSC-Specific Molecules and Were
were performed on 4-μm thick sections using standard methods.15,16 Enriched in a Distinctive Set of miRNAs and Proteins
Downloaded from http://ahajournals.org by on December 18, 2020

Capillary density was quantitated in isolectin-B4–stained sections.


To identify the molecular contents, which may be responsible
Interstitial collagen was quantified on images taken from the nonisch-
emic myocardium of picrosirius red-stained sections using ImagePro for the biological activity and regenerative capacity of EVs, we
Plus. Myocyte apoptosis was quantitated using TUNEL (terminal de- next performed global miRNA and proteomic profiling of iPSC-
oxynucleotidyl transferase dUTP nick-end labeling) assay.15 EV specimens. Among 282 miRNAs detected in iPSCs, 199
were also present in iPSC-EVs (Figure 2G), indicating efficient
Statistical Analysis transfer of these regulatory transcripts from the parental iPSCs
Data are expressed as mean±SEM or SD as indicated. The impact of
iPSC-EVs on the formation of CECs into capillary-like structures was to EVs. Importantly, miRNAs belonging to the miR-290-295
evaluated using the Mann–Whitney U test. The movement parameters cluster regulating pluripotency,19 including miR-292, -293, -294,
for CECs were compared using the Student t test. Morphometric and and miR-295, were abundant not only in the parental cells but
histological data were analyzed using 1-way ANOVA, whereas serial also in the iPSC-EVs (Online Tables I and II). Moreover, among
echocardiographic parameters were analyzed using 2-way (time and the miRNAs present in both iPSCs and iPSC-EVs, we found
group) ANOVA followed by Student t tests with the Bonferroni cor-
rection. Comparisons between 2 groups were performed by unpaired miR-19b, miR-20a, miR-126-3p, miR-130a-3p, miR-210-3p,
Student t test. P values <0.05 were considered statistically significant. and the longevimir cluster miR-17-92 reportedly involved in the
Statistical analyses were performed using the GraphPad Prism (ver- promotion of angiogenesis, adaptation to hypoxic stress, regula-
sion 4.03; GraphPad Software, Inc, La Jolla, CA) and SPSS software, tion of cell cycle, mammalian development, and aging.20
version 22.0 (IBM, Armonk, NY). Moreover, certain miRNAs were differentially expressed
in iPSCs and iPSC-EVs (Figure 2G; Online Tables I and
Results II). Indeed, 33 miRNAs were detected only in iPSC-EVs by
In Vitro Studies qPCR-based method (Figure 2G; Online Table III), which
iPSCs Expanded in Optimized Serum- and Feeder-Free suggests their high enrichment during EV production and
Conditions Maintained Pluripotency as a Source of release by iPSCs—a phenomenon suggested previously.21
Purified EVs MicroRNAs enriched in EVs, including let-7, miR-145, miR-
To obtain pure iPSC-EV specimens without contamination of 17-92 cluster, and ESC-specific miR-302a-5p, have been sug-
nanoparticles that originated from other cells or from serum gested to play a role in late developmental timing; regulation
present in the culture system,17 all iPSC clones were expanded of cell proliferation, differentiation, apoptosis, and mainte-
in optimized culture conditions in serum-free media without nance of self-renewal and pluripotency19,20,22; and may exert
feeder layer cells. Using a multi-instrumental approach, we possible beneficial effects on myocardial tissue. Moreover, the
confirmed the presence of a stable pluripotent phenotype in heatmap analysis indicated other signaling pathways poten-
the selected iPSC lines (Online Figure I). iPSCs expressed tially regulated by miRNA contents of iPSC-EVs, including
Adamiak et al   iPSC-EVs for Myocardial Repair   299
Downloaded from http://ahajournals.org by on December 18, 2020

Figure 1. Induced pluripotent stem cell (iPSC) characterization. A, Representative dot plots showing the expression of intracellular
markers of pluripotency in iPSCs. B–C, Representative images showing the morphology of iPSC colonies in serum- and feeder-
free culture and fluorescence staining for fetal alkaline phosphatase (PALP) activity (B), and expression of pluripotency markers
Oct3/4, Nanog, Sox2 (sex-determining region Y-box 2), and SSEA-1 (stage-specific embryonic antigen-1; C). Scale bar, 200 μm. D,
Representative histograms showing the antigenic phenotype of iPSCs by flow cytometry. The red-colored histograms represent samples
stained for specific surface antigens, whereas gray-colored ones correspond to respective unstained control samples. Data represent
mean±SD from 3 independent experiments. APC indicates allophycocyanin; BF, brightfield image; and PE, phycoerythrin.

Wnt, PI3K-Akt, and MAPK (mitogen-activated protein ki- Importantly, the number of proteins identified in challenging
nase) pathways, which are involved in the regulation of actin iPSC-EV samples and also in iPSC samples was highly re-
cytoskeleton, focal adhesion, and ECM-receptor interactions producible and consistent across all replicates, confirming the
(Online Figures IV and V). Moreover, additional examina- high quality of samples and proteomic procedures (Figure 3A).
tion by Ingenuity Pathway Analysis tool (Qiagen) identified Proteins within each group (iPSCs or iPSC-EVs) were consid-
several miRNAs abundant in iPSC-EVs, including miR-294, ered for analysis only when proteins were reliably identified
miR-16, miR-34, and miR-20, which may regulate VEGFA within the samples, ie, (1) in each replicate of the group and
(vascular endothelial growth factor A) signaling, thereby po- (2) in at least 2 of 3 replicates, on the basis of ≥2 unique pep-
tentially enhancing angiogenesis (Online Figure VI). tides. These strict analytic criteria were fulfilled by 4122 and
Next, we performed high-throughput global proteomic 1965 proteins detected in iPSCs and iPSC-EVs, respectively
analysis of iPSCs and iPSC-EVs using mass spectrometry. (Figure 3A). Because of the high number of common proteins
300  Circulation Research  January 19, 2018
Downloaded from http://ahajournals.org by on December 18, 2020

Figure 2. Characterization of induced pluripotent stem cell–derived extracellular vesicles (iPSC-EVs). A, Representative iPSC-EV
size distribution histogram by nanoparticle tracking analysis. The cumulative D50 parameter indicates that 50% of the population of
vesicles is <143 nm in diameter. B (left), representative atomic force microscopy (AFM) image of iPSC-derived EVs. Scale bar, 50 nm.
Right, 3D topography of individual vesicles. Scan area: 250×250 nm. C, The Young modulus (kPa) and adhesion (pN) values by AFM,
reflecting mechanical properties of iPSC-EVs. D, High-resolution flow cytometry. Left, Histogram showing the size distribution of mixed-
size synthetic beads (PS, fluorescently labeled polystyrene calibration beads detected in FITC (fluorescein isothiocyanate) channel; Si,
unlabeled silicone calibration beads). Right, Representative dot plots show the medium-angle light scatter (MALS) related to EV size
vs fluorescence intensity indicating expression of selected EV-specific (CD81) and iPSC-specific (SSEA-1 [stage-specific embryonic
antigen-1]) antigens on iPSC-EVs. E, Representative Western immunoblot confirming the presence of typical exosomal marker CD9 in
iPSC-EV specimens. F, Presence of mRNA transcripts for pluripotency-related markers in iPSCs and iPSC-EVs by real-time RT-PCR.
Data are shown as CT mean values. G, Venn diagram showing the number of miRNAs common and specific for iPSCs and iPSC-EVs. H,
Scatter plot of miRNA expression in iPSCs (x axis) and iPSC-EVs (y axis); each dot represents 1 transcript. Data represent mean±SD from
3 independent experiments. PE indicates phycoerythrin.

detected in both groups, confirming again the effective transfer Figure 3B through 3E and Online Figure VII. As expected,
of major molecular contents from the parental iPSCs to EVs, the proteins identified in iPSC-EVs were the most strongly
we focused our analysis on comparing the unique proteins iden- represented (by GO terms) in plasma membrane, cytosol, and
tified in all samples of a given group and none of the samples cytoskeleton but not in other organelle compartments not pres-
from the other groups. A total of 1536 differential proteins were ent in EV structure (Figure 3C). Importantly, the GO analysis
found to be present in iPSCs and 191 in iPSC-EVs (Figure 3A). of biological processes revealed an enrichment of differential
GO analysis for molecular function, cellular component, proteins involved in response to wound healing, regulation of
and biological processes related to the most apparent differ- cell differentiation, as well as organ, anatomic structure, and
ences between parental iPSCs and iPSC-EVs are shown in multicellular organism development (Figure 3D; Online Figure
Adamiak et al   iPSC-EVs for Myocardial Repair   301

A B
Group No. of all No. of No. of Common Differential iPSCs iPSC-derived EVs
sample proteins proteins proteins proteins in proteins in
identified in identified in identified in the group* the group†
the group the sample the sample
based on 2 or
Molecular function

% of proteins in the given GO term


more unique 30
peptides
25
iPSCs
20
1 6325 5412 4156 4122 1536
15
2 5551 4315
10
3 5561 4351
iPSC-EVs 5

1 4111 3329 2253 1965 191 0

2 2923 1838
3 3427 2308
*Proteins identified in at least 2 of 3 samples on the basis of two or more unique peptides.
†Proteins identified in all samples of a given group and none of the samples from the other group.

C Cellular component
% of proteins in the given GO term

16

14

12

10

D Biological process
% of proteins in the given GO term

18
Downloaded from http://ahajournals.org by on December 18, 2020

16
14
12
10
8
6
4
2
0

E 35 Molecular function
% of proteins in the given GO term

30

25

20

15

10

Figure 3. Global proteomic contents of induced pluripotent stem cells (iPSCs) and iPSC-derived extracellular vesicles (iPSC-EVs)
by mass spectrometry. A, The number of proteins identified in iPSCs and their EVs. The absolute values are shown individually for each
replicate (n=3) and as average for each examined group. B–D, Gene ontology (GO) analysis, including common proteins detected in
both iPSCs and iPSC-EVs, with focus on molecular function, cellular components, and biological processes. E, GO enrichment analysis
for differential expression of proteins in iPSCs and iPSC-EVs, with focus on molecular function. GO terms related to the most apparent
differences between iPSCs and iPSC-EVs are shown.
302  Circulation Research  January 19, 2018

VIIA). Importantly, several proteins involved in angiogenesis cytometry. Exposure to iPSC-EVs significantly decreased
signaling pathways (VEGF-C [vascular endothelial growth the incidence of staurosporine-induced apoptosis (38.1±4.7%
factor C], BMP-4 [bone morphogenetic protein-4], PDGFα versus 54.4±1.7% of cells in late apoptosis among CECs in-
[platelet-derived growth factor α], TDGF1 [teratocarcinoma- cubated with iPSC-EVs versus control-untreated cells, respec-
derived growth factor 1], CTGF [connective tissue growth tively; P<0.05; Figure 4D). Moreover, the number of viable
factor], and thrombospondin-1) were found within GO group cells was significantly greater in samples treated with iPSC-
guiding multicellular organism development (Online Figure EVs (29.3±5.3%) when compared with control CECs treated
VIIIA). Moreover, proteins stimulating cardiomyogenesis with staurosporine alone (13.4±0.7%). Importantly, there was
(BMP-4 and FGFs [fibroblast growth factors]); promoting no change in the number of cells undergoing necrosis in either
cardiac, endothelial, and smooth muscle cell proliferation iPSC-EV–treated or untreated samples (Figure 4D). These re-
(PDGFs, IGF-2 [insulin-like growth factor-2], and FGFs); and sults indicate cytoprotective and antiapoptotic effects of iPSC-
protecting cells against oxidative damage (hemopexin) were EVs on CECs in vitro, which may play an important role in
identified within GO group involved in signal transduction protecting ischemic myocytes in vivo.
(Online Figure VIIIB). Thus, proteomic data indicated that the
majority of iPSC-specific proteins is transferred from the pa- In Vivo Studies
rental iPSCs to iPSC-EVs and may suggest the impact of such Exclusions
protein contents on several biological processes in target cells, Four mice died in the perioperative period, and 3 mice died at
including the ones associated with tissue repair. 5, 19, and 29 days after intramyocardial injection. Eight mice
iPSC-EVs Improved CEC Function by Enhancing were excluded from the study because of cardiac tumor for-
Angiogenic Capacity, Migration, and Survival In Vitro mation in the iPSC-treated group, leaving a total of 9, 7, and
Increasing evidence indicates that angiogenesis is a major 12 mice in vehicle, iPSC, and iPSC-EV groups, respectively
mechanism responsible for the improvement in LV function (Online Table IV).
with cell therapy after ischemic myocardial injury. Because
Myocardial Infarct Size
our data identified several miRNAs and proteins within the
The infarct area fraction denotes the average area of scar tis-
iPSC-EVs, which may regulate target cell function, including
sue, expressed as a percentage of the LV area in 3 LV sections
proliferation, differentiation, survival, and angiogenesis, we
0.5 to 1.0 mm apart. The average infarct area fraction did not
investigated the impact of iPSC-EVs on selected functions of
differ significantly among the 3 groups (Online Figure X).
murine CECs in vitro, with emphasis on features potentially
important for heart repair. First, we confirmed that iPSC-EVs
Downloaded from http://ahajournals.org by on December 18, 2020

Intramyocardial Injection of iPSC-EVs Induced Greater


were internalized by CECs after only 1 hour of coincubation, Attenuation of LV Dysfunction
as shown by optical orthogonal sectioning in xy axis by fluo- Before coronary occlusion (baseline), all parameters of LV
rescent microscopy (Figure 4A). Compared with control-un- function were similar in groups I through III (Figure 5). At
treated cells, CECs treated with iPSC-EVs exhibited greater 48 hours after cell or EV transplantation (4 days after MI),
ability to form endotubules on Matrigel by quantitative analy- the degree of LV dysfunction was also similar among the 3
sis (Figure 4B; Online Figure IX; Online Movies I and II). groups (Figure 5B and 5C), indicating that the injuries sus-
These data suggest that iPSC-EVs are likely to exert proan- tained during ischemia/reperfusion and intramyocardial injec-
giogenic effects after myocardial injection in vivo. tion were similar in all groups. In vehicle-treated (group I)
Next, we addressed whether iPSC-EVs influence CEC mi- mice, there was further LV functional deterioration between
gratory capacity, which has been reported as a crucial functional 4 and 35 days after reperfusion. In contrast, both iPSC- and
property required in wound healing, tissue repair, including iPSC-EV–treated mice (groups II and III, respectively) exhib-
myocardial regeneration, as well as during new vessel forma- ited significantly improved global (Figure 5B through 5E) LV
tion and maturation.23 The quantitative analysis of recorded CEC function compared with group I at 35 days after MI. Mice in
trajectories, based on time-lapse monitoring of spontaneous un- groups II and III also showed enhanced regional myocardial
guided movement of single cells, showed greater speed of cell function in the infarct zone, as evidenced by a 10% (P<0.05)
displacement after iPSC-EV treatment compared with controls and 19% (P<0.01) greater systolic infarct wall thickness
(Figure 4C). This phenomenon was more pronounced and sta- (Figure 5A and 5F), respectively, compared with group I. The
tistically significant after prolonged EV treatment, when migra- preservation of LV function was significantly greater in hearts
tion was analyzed between 9 and 14 hours of CEC coincubation injected with iPSC-EVs compared with iPSC-injected hearts
with iPSC-EVs. These data indicate that iPSC-EVs enhance the (Figure 5B and 5C). In addition, mice in group III also exhib-
migratory capacity of CECs, which may augment new vessel ited smaller LV end-systolic volume compared with vehicle-
formation and maturation after iPSC-EV transplantation in vivo. treated mice (Figure 5E).
Finally, to examine the impact of iPSC-EVs on cell surviv-
al in cytotoxic conditions that often accompany tissue injury, Intramyocardial Delivery of iPSCs and iPSC-EVs Halted
we used staurosporine (1 μM), a cytotoxic agent commonly LV Remodeling and Hypertrophy
used to induce apoptosis, to challenge CECs before their incu- The echocardiographic measurements of LV remodel-
bation with iPSC-EVs. We measured cell death processes, in- ing at 35 days mirrored the observations on LV function.
cluding apoptosis and necrosis, after 24 hours of coincubation The LV end-diastolic diameter was significantly smaller
of staurosporine-challenged CECs with iPSC-EVs by flow in groups II and III compared with group I (Figure 6B).
Adamiak et al   iPSC-EVs for Myocardial Repair   303
Downloaded from http://ahajournals.org by on December 18, 2020

Figure 4. Impact of induced pluripotent stem cell–derived extracellular vesicles (iPSC-EVs) on cardiac endothelial cell (CEC)
function. A, Representative Z-stacked image showing uptake of fluorescently labeled iPSC-EVs by CECs. One set of orthogonal
slices is shown. Middle, right, and bottom represent XY, YZ, and XZ planes, respectively. YZ and XZ planes intersect according to the
crosshairs. Scale bar, 10 μm. B–D, Impact of iPSC-EVs on CEC functions: (B) angiogenic capacity on Matrigel; scale bar, 100 μm. Each
bar represents mean values±SD from 3 independent experiments (*P<0.1, **P<0.01, ***P<0.001); (C) migratory activity of untreated
CECs (control) and CECs treated with iPSC-EVs. Cell trajectories for each period are depicted as circular diagrams (axis scale in μm).
Selected migration parameters are shown on the graphs (bottom). Each bar represents mean values from 3 independent experiments
(*P<0.05); (D) representative dot plots showing the impact of treatment with iPSC-EVs on survival of CECs after exposure to cytotoxic
agent staurosporine. Data represent mean±SD from 3 independent experiments (*P<0.05). CME indicates coefficient of movement; PE,
phycoerythrin efficiency; and STS, staurosporine.

Also, compared with group I, the infarct wall thickness hypertrophy of the viable myocardium. Although the im-
in diastole was significantly greater both in groups II and provements in LV end-diastolic diameter and infarct wall
III (Figure 6C), indicating superior infarct repair. The thickness were numerically greater in iPSC-EV– compared
posterior LV wall thickness was smaller in groups II and with iPSC-treated hearts, the differences were not statisti-
III compared with group I (Figure 6D), indicating less cally significant.
304  Circulation Research  January 19, 2018

Figure 5. Assessment of left ventricular


(LV) systolic function. A, Representative
M-mode images from vehicle-, induced
pluripotent stem cell (iPSC)-, and iPSC-
derived extracellular vesicles (iPSC-
EV)–treated mice at 35 d after coronary
occlusion/reperfusion. Compared with
the vehicle-treated heart, both iPSC- and
iPSC-EV–treated hearts exhibit improved
wall motion, smaller LV cavity (D and E),
and thicker infarct wall (F). Transplantation
of iPSC-EVs resulted in greater
improvement in LV systolic function (B
and C). Data are mean±SEM. n=7 to 12
mice per group. BSL indicates baseline.
*P<0.05 vs vehicle at 35 d, #P<0.05 vs
iPSC group at 35 d.

Intramyocardial Delivery of iPSC-EVs Attenuated 53.1±1.6% of the infarct zone in groups I, II, and III, respec-
LV Hypertrophy tively (Figure 8A through 8D); therefore, the amount of vi-
To assess the impact of therapy on the myocardium, interstitial able myocardium in the infarct zone was, on average, 14% and
fibrosis was carefully quantitated. At 35 days after MI, inter- 18% greater in iPSC- and iPSC-EV–treated mice compared
stitial fibrosis was similar in vehicle- and iPSC-treated hearts with vehicle-treated mice, respectively (both P<0.05 versus
(Figure 6E and 6F). Although not statistically significant, in
Downloaded from http://ahajournals.org by on December 18, 2020

vehicle-treated group). However, there was no significant dif-


mice treated with iPSC-EVs, interstitial fibrosis was 11% less ference between iPSCs and iPSC-EVs groups.
compared with vehicle-treated hearts (Figure 6E and 6F). At In the infarct zone, the percentage of TUNEL-positive
35 days after MI, the echocardiographically estimated LV mass apoptotic cardiomyocyte nuclei was similar in groups I and II,
increased significantly in all groups compared with baseline whereas iPSC-EV treatment reduced apoptosis modestly yet
values. Compared with group I, the LV mass was 16% (P<0.05) significantly (Figure 8E and 8F). In the borderzone and non-
smaller in iPSC-EVs–treated mice (Figure 6G). Taken togeth- ischemic zone, compared with vehicle injection, iPSC injec-
er, these data indicate that injection of iPSC-EVs attenuates tion reduced the percentage of apoptotic myocytes minimally,
the adverse adaptations in surviving LV myocardium after MI. whereas iPSC-EV injection was associated with significant
Effects of iPSC and iPSC-EV Therapy on Myocardial reduction in myocyte apoptosis. These data indicate that in-
Capillary Density tramyocardial delivery of iPSC-EVs protects against myocyte
Because myocardial vascularity plays an important role in apoptosis during postinfarct remodeling.
cardiac repair post-MI, we assessed capillary density in the Intramyocardial Delivery of iPSCs Induced Teratoma
infarct zone, borderzone, and nonischemic zone separately. Formation
At 35 days after MI, compared with vehicle-treated hearts, Development of cardiac tumor was noted in a large number of
capillary numbers in nonischemic zone and infarct border- mice (8 of 15; 53%) in the iPSC-treated group. Gross morpho-
zone were greater in both iPSC- and iPSC-EV–treated hearts logical features, variegated tissue composition, and histologi-
(Figure 7). In the infarct zone, compared with vehicle treat- cal characteristics suggesting differentiation into derivatives
ment, iPSC-EV injection, but not iPSC injection, resulted in of all 3 germ layers identified these tumors as teratomas
significantly greater capillary numbers. These data indicate (Figure 8G; Online Figure XI). No such tumor was noted in
that intramyocardial delivery of both iPSCs and iPSC-EVs any mouse in iPSC-EV– or vehicle-injected groups.
promotes angiogenesis in infarct border zone and nonisch-
emic zone, whereas iPSC-EV therapy extends neovasculariza-
Discussion
tion in the infarct zone as well.
Although numerous strategies for cell-based cardiac repair
Effects on Viable Myocardium Within the Infarct Zone and have been tested in patients with ischemic heart disease, the
Myocyte Apoptosis benefits of such therapies are still only modest in clinical tri-
The effects of reparative therapies with regard to potential cel- als.1,24 Growing evidence indicates that paracrine effects of
lular replacement and myocyte salvage were assessed quan- EVs released by various cells can protect the endogenous
titatively. Myocytes constituted 45.1±1.2%, 51.4±2.7%, and myocardial tissue from cell death and other adverse effects,
Adamiak et al   iPSC-EVs for Myocardial Repair   305
Downloaded from http://ahajournals.org by on December 18, 2020

Figure 6. Assessment of left ventricular (LV) remodeling and hypertrophy. A, Representative Masson trichrome-stained myocardial
sections at 35 d after myocardial infarction (MI) show improved remodeling in induced pluripotent stem cell (iPSC)- and iPSC-derived
extracellular vesicles (iPSC-EV)–treated hearts. Scar tissue and viable myocardium are identified in blue and red, respectively. Scale
bar, 500 μm. Echocardiographically estimated LV end-diastolic diameter (B) was smaller in both iPSC- and iPSC-EV–treated groups
compared with the vehicle-treated group. LV infarct wall thickness in diastole (C) was greater and posterior wall thickness (D) was smaller
in both groups. E and F, Interstitial fibrosis in the viable myocardium was quantitated in picrosirius red-stained myocardial sections (E) at
35 d after MI and quantified (F). Scale bar, 50 μm. G, Echocardiographically estimated LV mass was smaller in iPSC-EV–treated hearts
compared with vehicle-treated hearts. Data are mean±SEM. n=7 to 12 mice per group. BSL indicates baseline. *P<0.05 vs vehicle at 35 d.

leading to improved remodeling and preserved function.8,25 parent cell, along with a distinct set of miRNAs and proteins;
The current results provide novel information in this regard. (2) iPSC-EVs enhance angiogenic capacity, migratory proper-
ties, and survival of heart-derived cells; (3) intramyocardial
Salient Findings transplantation of both iPSCs and iPSC-EVs after a reper-
We performed an extensive multidimensional evaluation of fused MI improves LV function, albeit iPSC-EVs render supe-
the molecular contents, biological effects, and regenerative rior benefits; (4) the in vitro effects of iPSC-EVs are translated
capacity of iPSC-EVs, consisting of both ectosomal and exo- in vivo with reduced myocyte apoptosis and enhanced angio-
somal fractions, on cardiac cell function in vitro. For the first genesis; and (5) iPSC injection is associated with tumor for-
time, the safety and cardiac reparative efficacy of iPSC-EVs mation, whereas iPSC-EVs seem safe. Together, these results
were directly compared with those of iPSCs in a model of indicate that injection of bioactive, cell-free, iPSC-EV speci-
reperfused MI in vivo. Our results show that (1) iPSCs release mens represents an effective and safe approach for cardiac re-
EVs that carry significant amounts of bioactive contents of the pair after ischemic injury.
306  Circulation Research  January 19, 2018

Figure 7. Impact on myocardial


capillary density. A, Representative
images of capillary profiles in the
infarct zone (IZ), border zone (BZ), and
nonischemic zone (NZ). B, Quantitative
myocardial capillary density. Data
represent mean±SEM. Scale bar, 50
μm. EV indicates extracellular vesicle;
and iPSC, induced pluripotent stem cell.
*P<0.05 vs vehicle.

Paracrine activities of transplanted cells have been widely as self-renewal and pluripotency.19 Moreover, the iPSC-EV–
implicated in tissue regeneration and heart repair. Indeed, our enriched miRNAs were accompanied with other transcripts
previous work suggests that adult bone marrow-derived cells common for iPSCs, including miR-290-295 embryonic cluster
improve heart function by increasing new vessel formation regulating pluripotency,19,33 as well as miR-19b, miR-20a, miR-
and myocardial perfusion because of growth factors released 126-3p, miR-130a-3p, miR-210-3p, and embryonic longevimir
by injected cells.26,27 Recent evidence also indicates that tiny cluster miR-17-92, which promote angiogenesis, adaptation
vesicles termed EVs carry various bioactive molecules that to hypoxic stress, regulation of cell cycle, mammalian devel-
may ultimately mediate the paracrine effects of stem cells. opment, and aging.20 These data suggest potential regulatory
Although the composition of EV cargo depends on the specif- effects of iPSC-EVs on survival, neovascularization, and pro-
ic cell type of origin, and influences the EV functional effects regenerative properties after the transfer of such miRNAs into
on target cells,28 iPSC-EVs have not been characterized in suf- cardiac cells.
ficient detail. Our current data constitute the first comprehen- Consistent with these findings at the miRNA level, we not
sive multi-instrumental analysis of cargo in murine iPSC-EVs only found ≈2000 proteins that were common for iPSC-EVs
and show that iPSC-EVs are enriched in numerous mRNA, and their parental cells but also ≈200 proteins, which were en-
Downloaded from http://ahajournals.org by on December 18, 2020

miRNA, and proteins that originate from parental iPSCs and riched in iPSC-EVs. The GO analysis identified sets of proteins
also possess a unique set of factors, which may be transferred enriched in iPSC-EVs, which are involved in response to ex-
to cardiac cells. The miRNAs and proteins in iPSC-EVs were ternal stimuli, wound healing, regulation of cell differentiation,
related not only to pluripotency but also to activation of cell as well as development of organs and multicellular organisms
proliferation, differentiation, and angiogenic activity. (Figure 3; Online Figure VIA). Importantly, several proteins in-
Importantly, we cultured iPSCs in feeder- and serum-free volved in angiogenesis and remodeling were identified as node
conditions in composition-defined medium, which are crucial proteins within the network (Online Figure VII). Together, the
factors for the quality and purity of iPSC-EVs. It has been current proteomic data identify key novel components of the
shown that the presence of any serum in EV donor cell culture iPSC-EV cargo that may exert reparative effects in vivo.
significantly increased the number of serum-derived particles The above findings also indicate that the contents of iP-
coisolated with the EV specimens, which may influence the re- SCs are efficiently transferred into EVs, thereby enabling
sults and data interpretation.29 Moreover, several laboratories, these vesicles as conveyors of iPSC molecular properties. The
including ours, have reported major impacts of different expan- potential role of EVs in transfer of proregenerative functions
sion media on the molecular composition and biological prop- to target cells was confirmed by increased angiogenic capac-
erties of EVs released by the same type of stem cell, suggesting ity, migratory ability, and resistance to apoptosis of CECs af-
the critical need for appropriate EV preparation and character- ter exposure to iPSC-EVs (Figure 4). These data also extend
ization before their administration.30,31 Thus, we selected serum- our previous observations on the transfer of bioactive cargo
free, fully defined medium for iPSC culture before iPSC-EV from human iPSC-EVs to primary cardiac mesenchymal
harvest, which simulated closely the GMP standards for cellular cells, thereby improving their proliferation, metabolic activ-
expansion for future application in humans. Our results confirm ity, as well as angiogenic and cardiomyogenic differentia-
that similar to iPSCs cultured in presence of serum,32 iPSCs ex- tion.3 Furthermore, these molecular findings provide plausible
panded under our defined serum-free conditions preserved plu- mechanisms underlying the iPSC-EV–induced benefits after
ripotent characteristics (Figure 1) and released iPSC-EVs with injection into the infarcted heart, which include (1) enhanced
normal morphology and antigenic phenotype (Figure 2). angiogenesis, which in turn leads to superior remodeling;
By performing global miRNA analysis, we found >200 (2) reduced apoptosis, which leads to greater preservation
regulatory miRNAs in iPSC-EV cargo, including 33 that of myocytes within the scar region; and (3) reduced fibrosis,
were enriched in the vesicles when compared with the donor which leads to improved remodeling and reduced LV mass.
iPSCs (Online Table I). The miRNAs enriched in iPSC-EVs However, based on the size criteria, iPSC-EVs used in this
included the miR-145, let-7 family, and miR-302a-5p that are study contained both ectosomes and exosomes, described
known to regulate cell proliferation, differentiation, as well as distinct EV subpopulations in terms of biogenesis, size,
Adamiak et al   iPSC-EVs for Myocardial Repair   307
Downloaded from http://ahajournals.org by on December 18, 2020

Figure 8. Impact on cardiomyocyte salvage and neoplastic growth. Viable myocyte area fraction in the infarct zone (IZ). A–C,
representative examples of the infarct scar area in Masson trichrome-stained vehicle-treated (A), induced pluripotent stem cell
(iPSC)-treated (B), and iPSC-EV–treated (C) hearts. Scale bar, 50 μm. Quantitative data are presented in (D). Data are mean±SEM.
n=7 to 12 mice per group. *P<0.05 vs vehicle. Myocyte apoptosis. E, Representative images from the infarct borderzone after TUNEL
(terminal deoxynucleotidyl transferase dUTP nick-end labeling) staining at 35 d after myocardial infarction. Apoptotic nuclei (white
arrows) are visualized by the green fluorescence. DAPI (4’,6-diamidino-2-phenylindole dihydrochloride) staining identifies nuclei
in blue. Cardiac myocytes are positive for α-sarcomeric actin (red). Scale bar, 50 μm. Quantitative data are presented in F. Data
represent mean±SEM. *P<0.05 vs vehicle, #P<0.05 vs iPSCs. BZ indicates borderzone; IZ, infarct zone; and NZ, (Continued )
308  Circulation Research  January 19, 2018

Figure 8 Continued. nonischemic zone. G, representative gross morphologies of tumors from 2 iPSC-injected hearts (top);
photomicrographs from Masson trichrome-stained myocardial sections showing the variegated tissue composition of tumors (middle;
scale bar, 200 μm); and differentiation into ectodermal (neuroectoderm with pigment granules [arrows]; lower left), mesodermal (cartilage;
lower middle), and endodermal (respiratory epithelium with cilia [arrows]; lower right) lineages in hematoxylin and eosin–stained sections
from hearts harboring tumors. Scale bar, 50 μm.

molecular contents, and function.34 The potential differences is a well-known phenomenon related to the pluripotent state
in bioactive molecular contents between these 2 fractions and of these cells and has been observed in several prior stud-
the mechanistic implications thereof with regard to heart re- ies.11,38,39 This phenomenon is currently one of the major
pair by each fraction separately remain to be explored. hurdles for therapeutic application of iPSCs for tissue re-
Although both iPSCs and EVs have been used for car- generation in humans. Our findings show that even though
diac repair, each has unique advantages and disadvantages. the injection of parental iPSCs remains hazardous, superior
Therefore, we felt it was important to directly compare the reparative benefits may be achieved safely with the injection
outcomes of cardiac repair with iPSCs and iPSC-EVs in vivo. of EVs derived from those same cells.
Although both improved LV function, the benefits of iPSC-
EVs were more pronounced. In addition, the improvement in Conclusions
LV remodeling was superior with iPSC-EVs. This was evi- The current findings provide comprehensive evidence that iP-
dent in smaller LV end-systolic volume and LV mass in iPSC- SC-EVs represent bioactive specimens that carry unique pay-
EV–treated mice. Although iPSC-EV injection also reduced loads of molecules released by donor iPSCs, which may be
interstitial fibrosis by 11%, this did not reach statistical sig- transferred to cardiac cells resulting in enhancement of biologi-
nificance. Furthermore, iPSC-EV injection enhanced capillary cal functions. Further, the first systematic comparison of iPSCs
density in the infarct zone, which may enhance the quality of and iPSC-EVs in vivo reveals superior efficacy of iPSC-EVs
for cytoprotection, vascularization, and cardiac repair. Finally,
scar and further improve remodeling during longer follow-up.
the absence of tumor formation in the iPSC-EV group advances
This effect may directly relate to the transference of proangio-
the potential candidacy of the same for safe and effective heart
genic properties by iPSC-EVs to host myocardial cells.
repair after ischemic myocardial injury in humans.
The reduced incidence of myocyte apoptosis in hearts in-
jected with iPSC-EVs constitutes another important observa-
tion. These findings are consistent with the in vitro findings Acknowledgments
We thank Dr Grazyna Drabik, MD, from the Department of
of greater survival of CECs after incubation with iPSC-EVs.
Transplantation, Polish-American Children’s Hospital in Krakow for
Similar cytoprotective effects of iPSC-EVs were noted in a pre-
Downloaded from http://ahajournals.org by on December 18, 2020

assistance with pathological analysis of tumors.


vious study by Wang et al,35 However, in that study, exosomes
were injected immediately after the coronary occlusion and Sources of Funding
before reperfusion was achieved. The reduction of apoptosis This work was supported, in part, by the National Institutes of
with exosome therapy was noted after 24 hours. The authors Health grant R01 HL-117730 (B. Dawn), 2013/10/E/NZ3/00750 and
reported reduced myocardial injury, which was explained pre- 2015/16/W/NZ4/00071 grants from National Science Center (E.K.
dominantly by cytoprotective effects mediated by miR-21 and Zuba-Surma), and TEAM-2012/9-6 grant from the Foundation for
Polish Science (E.K. Zuba-Surma). The Faculty of Biochemistry,
miR-210 transferred from exosomes to cardiac cells.35 Our re- Biophysics, and Biotechnology at the Jagiellonian University,
sults show, for the first time, that iPSC-EVs confer cytoprotec- Krakow, Poland, is a partner of the Leading National Research Center
tive benefits even when administered after reperfusion injury (KNOW) supported by the Ministry of Science and Higher Education.
has set in and these benefits persist during longer follow-up.
Although several miRNAs have been implicated in cytopro- Disclosures
tection, miRNAs from the miR-17-92 cluster (eg, miR-19a, None.
miR-19b, and miR-20a) noted in high concentration in iPSC-
EVs may be responsible for this observation.36 Moreover, the References
miR-17-92 cluster also enhances angiogenesis,37 which may 1. Afzal MR, Samanta A, Shah ZI, Jeevanantham V, Abdel-Latif A, Zuba-
contribute toward improvement in heart function after iPSC- Surma EK, Dawn B. Adult bone marrow cell therapy for ischemic heart
disease: evidence and insights from randomized controlled trials. Circ Res.
EV injection. Consistently, our proteomic data also indicated 2015;117:558–575. doi: 10.1161/CIRCRESAHA.114.304792.
the enrichment of several proangiogenic molecules in iPSC- 2. Chimenti I, Smith RR, Li TS, Gerstenblith G, Messina E, Giacomello A,
EVs, including BMP-4, PDGFα, TDGF1, thrombospondin-1, Marbán E. Relative roles of direct regeneration versus paracrine effects of
human cardiosphere-derived cells transplanted into infarcted mice. Circ
and VEGF-C. This enrichment of the molecular contents of
Res. 2010;106:971–980. doi: 10.1161/CIRCRESAHA.109.210682.
iPSC-EVs with proangiogenic and cytoprotective agents may 3. Bobis-Wozowicz S, Kmiotek K, Sekula M, Kedracka-Krok S, Kamycka
explain the beneficial effects observed in vivo. E, Adamiak M, Jankowska U, Madetko-Talowska A, Sarna M, Bik-
Finally, the direct comparison of iPSCs and iPSC-EVs Multanowski M, Kolcz J, Boruczkowski D, Madeja Z, Dawn B, Zuba-
Surma EK. Human induced pluripotent stem cell-derived microvesicles
also brings to light a vitally important finding with regard transmit RNAs and proteins to recipient mature heart cells modulating
to safety of regenerative therapy. Although the follow-up cell fate and behavior. Stem Cells. 2015;33:2748–2761. doi: 10.1002/
duration was only 6 weeks, 8 of 15 mice injected with iP- stem.2078.
4. Théry C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis
SCs developed teratomas within various cardiac structures.
and function. Nat Rev Immunol. 2002;2:569–579. doi: 10.1038/nri855.
No such neoplastic transformation was noted in the other 2 5. Mulcahy LA, Pink RC, Carter DR. Routes and mechanisms of extracellu-
groups. Teratoma formation by iPSCs after injection in vivo lar vesicle uptake. J Extracell Vesicles. 2014;3. doi: 10.3402/jev.v3.24641.
Adamiak et al   iPSC-EVs for Myocardial Repair   309

6. György B, Szabó TG, Pásztói M, Pál Z, Misják P, Aradi B, László V, 22. Cordes KR, Sheehy NT, White MP, Berry EC, Morton SU, Muth AN, Lee
Pállinger E, Pap E, Kittel A, Nagy G, Falus A, Buzás EI. Membrane ves- TH, Miano JM, Ivey KN, Srivastava D. miR-145 and miR-143 regulate
icles, current state-of-the-art: emerging role of extracellular vesicles. Cell smooth muscle cell fate and plasticity. Nature. 2009;460:705–710. doi:
Mol Life Sci. 2011;68:2667–2688. doi: 10.1007/s00018-011-0689-3. 10.1038/nature08195.
7. Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötvall JO. 23. Tahara N, Brush M, Kawakami Y. Cell migration during heart regeneration
Exosome-mediated transfer of mRNAs and microRNAs is a novel mecha- in zebrafish. Dev Dyn. 2016;245:774–787. doi: 10.1002/dvdy.24411.
nism of genetic exchange between cells. Nat Cell Biol. 2007;9:654–659. 24. Jeevanantham V, Butler M, Saad A, Abdel-Latif A, Zuba-Surma EK,
doi: 10.1038/ncb1596. Dawn B. Adult bone marrow cell therapy improves survival and in-
8. Barile L, Lionetti V, Cervio E, Matteucci M, Gherghiceanu M, Popescu duces long-term improvement in cardiac parameters: a systematic re-
LM, Torre T, Siclari F, Moccetti T, Vassalli G. Extracellular vesicles view and meta-analysis. Circulation. 2012;126:551–568. doi: 10.1161/
from human cardiac progenitor cells inhibit cardiomyocyte apoptosis and CIRCULATIONAHA.111.086074.
improve cardiac function after myocardial infarction. Cardiovasc Res. 25. Lai RC, Arslan F, Lee MM, Sze NS, Choo A, Chen TS, Salto-Tellez M,
2014;103:530–541. doi: 10.1093/cvr/cvu167. Timmers L, Lee CN, El Oakley RM, Pasterkamp G, de Kleijn DP, Lim
9. Cantaluppi V, Medica D, Mannari C, Stiaccini G, Figliolini F, Dellepiane SK. Exosome secreted by MSC reduces myocardial ischemia/reperfusion
S, Quercia AD, Migliori M, Panichi V, Giovannini L, Bruno S, Tetta C, injury. Stem Cell Res. 2010;4:214–222. doi: 10.1016/j.scr.2009.12.003.
Biancone L, Camussi G. Endothelial progenitor cell-derived extracel- 26. Zuba-Surma EK, Guo Y, Taher H, Sanganalmath SK, Hunt G, Vincent
lular vesicles protect from complement-mediated mesangial injury in RJ, Kucia M, Abdel-Latif A, Tang XL, Ratajczak MZ, Dawn B, Bolli R.
experimental anti-Thy1.1 glomerulonephritis. Nephrol Dial Transplant. Transplantation of expanded bone marrow-derived very small embryonic-
2015;30:410–422. doi: 10.1093/ndt/gfu364. like stem cells (VSEL-SCs) improves left ventricular function and remod-
10. Collino F, Bruno S, Incarnato D, Dettori D, Neri F, Provero P, Pomatto elling after myocardial infarction. J Cell Mol Med. 2011;15:1319–1328.
M, Oliviero S, Tetta C, Quesenberry PJ, Camussi G. AKI recovery in- doi: 10.1111/j.1582-4934.2010.01126.x.
duced by mesenchymal stromal cell-derived extracellular vesicles carry- 27. Zuba-Surma EK, Wojakowski W, Ratajczak MZ, Dawn B. Very small em-
ing microRNAs. J Am Soc Nephrol. 2015;26:2349–2360. doi: 10.1681/ bryonic-like stem cells: biology and therapeutic potential for heart repair.
ASN.2014070710. Antioxid Redox Signal. 2011;15:1821–1834. doi: 10.1089/ars.2010.3817.
11. Gutierrez-Aranda I, Ramos-Mejia V, Bueno C, Munoz-Lopez M, Real PJ, 28. Xu R, Greening DW, Zhu HJ, Takahashi N, Simpson RJ. Extracellular
Mácia A, Sanchez L, Ligero G, Garcia-Parez JL, Menendez P. Human vesicle isolation and characterization: toward clinical application. J Clin
induced pluripotent stem cells develop teratoma more efficiently and faster Invest. 2016;126:1152–1162. doi: 10.1172/JCI81129.
than human embryonic stem cells regardless the site of injection. Stem 29. Inal JM, Kosgodage U, Azam S, Stratton D, Antwi-Baffour S, Lange
Cells. 2010;28:1568–1570. doi: 10.1002/stem.471. S. Blood/plasma secretome and microvesicles. Biochim Biophys Acta.
12. Théry C, Amigorena S, Raposo G, Clayton A. Isolation and characteriza- 2013;1834:2317–2325. doi: 10.1016/j.bbapap.2013.04.005.
tion of exosomes from cell culture supernatants and biological fluids. Curr 30. Bobis-Wozowicz S, Kmiotek K, Kania K, Karnas E, Labedz-Maslowska
Protoc Cell Biol. 2006;chapter 3:unit 3.22. doi: 10.1002/0471143030. A, Sekula M, Kedracka-Krok S, Kolcz J, Boruczkowski D, Madeja Z,
cb0322s30. Zuba-Surma EK. Diverse impact of xeno-free conditions on biological
13. Wiśniewski JR, Zougman A, Nagaraj N, Mann M. Universal sample prep- and regenerative properties of hUC-MSCs and their extracellular vesicles.
aration method for proteome analysis. Nat Methods. 2009;6:359–362. doi: J Mol Med (Berl). 2017;95:205–220. doi: 10.1007/s00109-016-1471-7.
10.1038/nmeth.1322. 31. Gardiner C, Di Vizio D, Sahoo S, Théry C, Witwer KW, Wauben M, Hill
14. Käll L, Canterbury JD, Weston J, Noble WS, MacCoss MJ. Semi-
AF. Techniques used for the isolation and characterization of extracellular
Downloaded from http://ahajournals.org by on December 18, 2020

supervised learning for peptide identification from shotgun proteomics vesicles: results of a worldwide survey. J Extracell Vesicles. 2016;5:32945.
datasets. Nat Methods. 2007;4:923–925. doi: 10.1038/nmeth1113. 32. Zhou J, Ghoroghi S, Benito-Martin A, Wu H, Unachukwu UJ, Einbond
15. Zhao L, Cheng G, Jin R, et al. Deletion of interleukin-6 attenuates
LS, Guariglia S, Peinado H, Redenti S. Characterization of induced plurip-
pressure overload-induced left ventricular hypertrophy and dysfunc- otent stem cell microvesicle genesis, morphology and pluripotent content.
tion. Circ Res. 2016;118:1918–1929. doi: 10.1161/CIRCRESAHA. Sci Rep. 2016;6:19743. doi: 10.1038/srep19743.
116.308688. 33. Lichner Z, Páll E, Kerekes A, Pállinger E, Maraghechi P, Bosze Z, Gócza
16. Dawn B, Tiwari S, Kucia MJ, Zuba-Surma EK, Guo Y, Sanganalmath SK, E. The miR-290-295 cluster promotes pluripotency maintenance by
Abdel-Latif A, Hunt G, Vincent RJ, Taher H, Reed NJ, Ratajczak MZ, regulating cell cycle phase distribution in mouse embryonic stem cells.
Bolli R. Transplantation of bone marrow-derived very small embryonic- Differentiation. 2011;81:11–24. doi: 10.1016/j.diff.2010.08.002.
like stem cells attenuates left ventricular dysfunction and remodeling af- 34. Cocucci E, Meldolesi J. Ectosomes and exosomes: shedding the confusion
ter myocardial infarction. Stem Cells. 2008;26:1646–1655. doi: 10.1634/ between extracellular vesicles. Trends Cell Biol. 2015;25:364–372. doi:
stemcells.2007-0715. 10.1016/j.tcb.2015.01.004.
17. Shelke GV, Lasser C, Gho YS, Lotvall J. Importance of exosome depletion 35. Wang Y, Zhang L, Li Y, Chen L, Wang X, Guo W, Zhang X, Qin G, He
protocols to eliminate functional and RNA-containing extracellular vesi- SH, Zimmerman A, Liu Y, Kim IM, Weintraub NL, Tang Y. Exosomes/
cles from fetal bovine serum. J Extracell Vesicles. 2014;3. doi: 10.3402/ microvesicles from induced pluripotent stem cells deliver cardioprotective
jev.v3.24783. miRNAs and prevent cardiomyocyte apoptosis in the ischemic myocar-
18. Lötvall J, Hill AF, Hochberg F, Buzás EI, Di Vizio D, Gardiner C, Gho YS, dium. Int J Cardiol. 2015;192:61–69. doi: 10.1016/j.ijcard.2015.05.020.
Kurochkin IV, Mathivanan S, Quesenberry P, Sahoo S, Tahara H, Wauben 36. Sylvestre Y, De Guire V, Querido E, Mukhopadhyay UK, Bourdeau

MH, Witwer KW, Théry C. Minimal experimental requirements for defini- V, Major F, Ferbeyre G, Chartrand P. An E2F/miR-20a autoregulatory
tion of extracellular vesicles and their functions: a position statement from feedback loop. J Biol Chem. 2007;282:2135–2143. doi: 10.1074/jbc.
the International Society for Extracellular Vesicles. J Extracell Vesicles. M608939200.
2014;3:26913. 37. Chamorro-Jorganes A, Lee MY, Araldi E, Landskroner-Eiger S,

19. Gruber AJ, Grandy WA, Balwierz PJ, Dimitrova YA, Pachkov M, Ciaudo Fernández-Fuertes M, Sahraei M, Quiles Del Rey M, van Solingen C, Yu
C, Nimwegen Ev, Zavolan M. Embryonic stem cell-specific microRNAs J, Fernández-Hernando C, Sessa WC, Suárez Y. VEGF-induced expres-
contribute to pluripotency by inhibiting regulators of multiple differentia- sion of miR-17-92 cluster in endothelial cells is mediated by ERK/ELK1
tion pathways. Nucleic Acids Res. 2014;42:9313–9326. doi: 10.1093/nar/ activation and regulates angiogenesis. Circ Res. 2016;118:38–47. doi:
gku544. 10.1161/CIRCRESAHA.115.307408.
20. Mendell JT. miRiad roles for the miR-17-92 cluster in development and 38. Liu Z, Tang Y, Lü S, Zhou J, Du Z, Duan C, Li Z, Wang C. The tumouri-
disease. Cell. 2008;133:217–222. doi: 10.1016/j.cell.2008.04.001. genicity of iPS cells and their differentiated derivates. J Cell Mol Med.
21. Guduric-Fuchs J, O’Connor A, Camp B, O’Neill CL, Medina RJ, Simpson 2013;17:782–791. doi: 10.1111/jcmm.12062.
DA. Selective extracellular vesicle-mediated export of an overlapping set 39. Kamada M, Mitsui Y, Matsuo T, Takahashi T. Reversible transformation
of microRNAs from multiple cell types. BMC Genomics. 2012;13:357. and de-differentiation of human cells derived from induced pluripotent stem
doi: 10.1186/1471-2164-13-357. cell teratomas. Hum Cell. 2016;29:1–9. doi: 10.1007/s13577-015-0119-1.

You might also like