Advanced Drug Delivery Reviews: Jin Han, Jooyeon Park, Byung-Soo Kim

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Advanced Drug Delivery Reviews 95 (2015) 15–28

Contents lists available at ScienceDirect

Advanced Drug Delivery Reviews

journal homepage: www.elsevier.com/locate/addr

Integration of mesenchymal stem cells with nanobiomaterials for the


repair of myocardial infarction☆
Jin Han a,1, Jooyeon Park a,1, Byung-Soo Kim a,b,⁎
a
School of Chemical and Biological Engineering, Seoul National University, Seoul 151-744, Republic of Korea
b
Interdisciplinary Program of Bioengineering, Bio-MAX Institute, Institute of Chemical Processes, Seoul National University, Seoul 151-744, Republic of Korea

a r t i c l e i n f o a b s t r a c t

Article history: The integration of nanobiomaterials with stem cells represents a promising strategy for the treatment of myocar-
Received 1 June 2015 dial infarction. While stem cells and nanobiomaterials each demonstrated partial success in cardiac repair indi-
Received in revised form 27 August 2015 vidually, the therapeutic efficacy of the clinical settings for each of these has been low. Hence, a combination of
Accepted 10 September 2015
nanobiomaterials with stem cells is vigorously studied to create synergistic effects for treating myocardial infarc-
Available online 24 September 2015
tion. To date, various types of nanomaterials have been incorporated with stem cells to control cell fate, modulate
Keywords:
the therapeutic behavior of stem cells, and make them more suitable for cardiac repair. Here, we review the cur-
Stem cells rent stem cell therapies for cardiac repair and describe the combinatorial approaches of using nanobiomaterials
Biomaterials and stem cells to improve therapeutic efficacy for the treatment of myocardial infarction.
Myocardial infarction © 2015 Elsevier B.V. All rights reserved.
Nanomaterials
Regenerative medicine
Tissue engineering

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
2. Mesenchymal stem cells (MSCs) for the treatment of myocardial infarction. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
2.1. Differentiation of MSCs into cardiac lineage cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
2.2. Paracrine factor secretion of MSCs for cardiac repair . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
3. Nanobiomaterial-incorporated stem cell therapy for cardiac repair. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
3.1. Therapeutic molecule delivery systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
3.2. Cell delivery vehicles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
3.3. Nanotopographical cues of nanobiomaterials . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
3.4. Electrically conductive nanobiomaterials . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 22
3.5. Intrinsic chemistry of nanobiomaterials . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
4. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25

Abbreviations: MI, myocardial infarction; MSC, mesenchymal stem cell; ESC, embryonic stem cell; iPSC, induced pluripotent stem cell; CSC, cardiac stem cell; PDGF-BB, platelet-derived
growth factor-BB; PlGF, placental growth factor; VEGF, vascular endothelial growth factor; SDF-1, stromal cell-derived factor 1; IGF-1, insulin growth factor 1; Cx43, connexin 43; MNC,
mononuclear cell; CMC, cardiomyocytes; ASC, adipose-derived stem cell; PLCL, poly(lactide-co-ε-caprolactone); PCL, polycaprolactone; PEI, polyethylenimine; PEG, polyethylene glycol;
BSA, bovine serum albumin; PVA, polyvinyl alcohol.
☆ This review is part of the Advanced Drug Delivery Reviews theme issue on "Multifunctional Nanodevices and Nanobots for Bioimaging, Cancer Diagnosis/Therapy, Stem Cell Therapy,
and Regenerative Medicine".
⁎ Corresponding author at: School of Chemical and Biological Engineering, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 151-744, Republic of Korea. Tel.: +82 2 880 1875;
fax: +82 2 2297 0838.
E-mail address: byungskim@snu.ac.kr (B.-S. Kim).
1
These authors contributed equally.

http://dx.doi.org/10.1016/j.addr.2015.09.002
0169-409X/© 2015 Elsevier B.V. All rights reserved.
16 J. Han et al. / Advanced Drug Delivery Reviews 95 (2015) 15–28

1. Introduction 2. Mesenchymal stem cells (MSCs) for the treatment of


myocardial infarction
Cardiovascular diseases remain one of the leading causes of death
worldwide [1,2]. Over 80 million individuals in United States have one A number of preclinical and clinical studies have been performed
or more types of cardiovascular diseases [2]. Myocardial infarction using stem cells to repair damaged hearts [15,18,21,52,53]. Most of
(MI), in particular, serves as the major contributor to heart failure and these cells, including MSCs, ESCs, iPSCs, and CSCs, improved cardiac
cardiac-associated disorders [1,3,4]. Previously, thrombolytic treat- function, suggesting that the implantation of stem cells is a promising
ments or surgical coronary artery bypass were introduced to treat MI; therapeutic approach for MI repair. ESCs and iPSCs represent ideal cell
however, these therapies merely promoted cardiac tissue regeneration sources for MI repair due to their pluripotency. ESCs can differentiate
[5–7]. MI is characterized by reduced blood supply to the heart, which into functional cardiomyocytes when implanted into the injured myo-
results in cardiac cell death, tissue damage, ventricular malfunction, cardium [16,17]. Differentiation of ESCs at the lesion site further restores
and heart failure [4]. Hence, successful treatments for MI depend on sal- contractile function of the heart and attenuates cardiac remodeling [15].
vaging dying cells, recovering blood vessels, and regenerating cardiac Similarly, the implantation of iPSCs into the injured myocardium results
tissues [4,7]. In other words, cardiac tissue engineering approaches in cell engraftment and differentiation into cardiomyocytes, which leads
that can prevent cell death and regenerate cardiac tissues are required to enhanced cardiac function [18–20]. Despite their therapeutic poten-
for effective treatment of MI. tial, ESCs pose ethical concerns and potential teratogenic risks while
Recently, significant advances have been made in the use of stem the generation efficiency of iPSCs is still very low [54,55]. As an alterna-
cells for the treatment of MI [8–10] and a number of clinical trials tive, CSCs have attracted attention as a cell source with great cardiac dif-
with stem cells showed encouraging results with cardiac function ferentiation potential which can attribute to the repair of the damaged
restoration. Stem cells have regenerative potentials to replace the heart. CSCs are c-kit-positive heart-resident stem cells that can differen-
dead cardiomyocytes, salvage dying cells, and induce angiogenesis tiate into cardiac lineage cells [55]. However, an extremely small
at the infarcted region [7]. A number of different cell types, including amount of CSCs within a heart and the difficulties in obtaining these
mesenchymal stem cells (MSCs) [11–14], emb ryonic stem cells cells limit their clinical applications despite their high therapeutic effi-
(ESCs) [15–17], induced pluripotent stem cells (iPSCs) [18–20], and cacy [55]. Meanwhile, MSCs have been extensively investigated as the
cardiac stem cells (CSCs) [21], all showed salutary effects in repairing optimal cell source for MI. The cardiac differentiation potential of
injured myocardium. Among the various cell types, MSCs represent a MSCs is comparatively limited compared to ESCs, iPSCs, and CSCs. How-
reliable source for cell therapy as they have shown safe and promis- ever, MSCs can easily be isolated from patients, expanded ex vivo, and
ing results in long-term clinical trials [12,14,22]. Upon implantation utilized in an autologous manner [7]. Additionally, the secretion of ther-
into the infarcted heart, a fraction of MSCs differentiates into func- apeutic paracrine molecules from MSCs, including various types of
tional cardiomyocytes and replaces the damaged cardiac tissues to growth factors and cytokines, is known to repair the injured myocardi-
improve heart function [23–25]. Additionally, MSCs secrete various um [1,56–60]. The therapeutic efficacy of MSCs has been exhibited in a
therapeutic paracrine molecules for cardiac cell protection, angio- number of clinical studies. The POSEIDON trial demonstrated that the
genesis, and immune modulation for cardiac repair [1,26–28]. How- implantation of autologous or allogenic human MSCs can improve car-
ever, low cell survival, engraftment, and differentiation of stem cells diac function after MI [52]. Additionally, the APOLLO trial showed en-
after implantation impede the therapeutic effect of the stem cells. hancements in cardiac function, perfusion, and neovasculogenesis
Hence, other treatment options are vigorously investigated for better after the transplantation of adipose-derived stem cells [53]. Various
cardiac repair. other trials, including the ANGEL, ATHENA, and MyStromal Cell, are cur-
Nanobiomaterial-based treatments for MI recently drew significant rently undergoing clinical phase trials for chronic myocardial ischemia
attention as new therapeutic approaches for cardiac repair [29,30]. [8]. In the following sections, we review the two major therapeutic
Nanobiomaterials represent natural or synthetic biocompatible mate- mechanisms of MSC therapy for MI, namely, (1) differentiation of
rials with nanoscale characteristics, which further provide exclusive MSCs into cardiac lineage and (2) paracrine factor secretion of MSCs
physicochemical properties such as nanoscale size, nanotopographical (Fig. 1).
cues, electrical conductivity, or distinct chemical features. Treatments
that utilize nanobiomaterials, including nanoparticles, nanofibrous 2.1. Differentiation of MSCs into cardiac lineage cells
scaffolds, self-assembling peptides, and many others, showed signifi-
cant effects in repairing injured myocardium [29]. A number of Direct differentiation of the implanted MSCs into cardiac lineage
nanobiomaterials were previously used to deliver therapeutic mole- cells was originally proposed as the principal mechanism of stem cell
cules including proteins, drugs, and genes for cardiac repair [31–35]. Ad- therapy for MI [7]. Yoon et al. showed that MSCs implanted to the in-
ditionally, biomaterials utilized as cell carriers have improved the farcted myocardium differentiated into cardiomyocytes, endothelial
therapeutic efficacy of cells for the treatment of MI [36,37]. More recent- cells, and smooth muscle cells, and consequently improved cardiac
ly, nanobiomaterials that can modulate cell functions have provided a function [61]. Differentiation of adipose-derived and cortical bone-
new window for nanobiomaterial-incorporated cell therapy for MI, derived stem cells into cardiomyocytes and vascular cells was observed
and materials with unique topographical, electrical, or chemical charac- after in vivo implantation [56,62]. However, the fraction of MSCs that
teristics have been extensively studied to create synergistic effects with differentiated into cardiomyocytes after in vivo implantation is relative-
cell therapy. Overall, the combination of conventional stem cell therapy ly low. Hence, many attempts were made to differentiate MSCs toward
and nanobiomaterials propose a great therapeutic option for treating MI cardiac lineages in vitro prior to in vivo implantation, with the help of
[38–51]. chemicals, small molecules, growth factors, and co-culture with cardiac
Here, we review the current status of tissue regenerative approaches cells, to improve the therapeutic efficacy of MSCs [63]. To differentiate
for treating MI, particularly stem cell therapy, nanobiomaterial-based MSCs into cardiomyocytes using chemicals, 5-azacytidine has been
biomolecule delivery, and nanobiomaterial-integrated stem cell thera- widely utilized as the key chemical facilitator [64,65]. It was also report-
py. We first review the mechanisms and recent advances in stem cell ed that the fluid shear stress in combination with 5-azacytidine treat-
therapy for MI. Next, we introduce nanobiomaterials with the potential ment further enhanced cardiomyogenic differentiation of MSCs [66].
to efficiently deliver therapeutic molecules or cells for the treatment of In addition to 5-azacytidine, small molecules such as phorbol myristate
MI. Finally, we discuss biomaterials with unique topographical, electri- acetate can also promote cardiomyogenic differentiation of MSCs, and
cal, or chemical characteristics that can improve the therapeutic efficacy stimulate electromechanical integration of MSCs with resident
of stem cells for cardiac repair. cardiomyocytes [67]. Bartunek et al. further showed that pretreatment
J. Han et al. / Advanced Drug Delivery Reviews 95 (2015) 15–28 17

Fig. 1. Therapeutic mechanisms involved in stem cell therapy for the treatment of MI. Stem cells can either differentiate into cardiomyocytes or secrete therapeutic paracrine molecules to
salvage and regenerate heart tissue.

of MSCs with cardiomyogenic growth factors prior to implantation stimulates their paracrine molecule secretion, but also results in their
improves cardiac lineage differentiation of MSCs in vivo and en- enhanced survival in the infarcted myocardium [26].
hances the functional recovery of a chronically infarcted myocardi- These observations regarding the cardiac differentiation capability
um [68]. The co-culture of MSCs with cardiac cells also promotes and paracrine action of MSCs prompted the development of new thera-
cardiomyogenesis of MSCs, which can be further promoted by peutic strategies that exert synergistic effects with these cells. Recently,
pretreating the cardiac cells with iron oxide nanoparticles [51] or nanobiomaterials that improve the cardiac differentiation and harness
by the alignment of the cells [69]. Once implanted into the damaged the paracrine action of MSCs have gained much attention. The advances
heart, MSC-differentiated cardiomyocytes can reduce fibrosis, im- in nanobiomaterial-integrated stem cell therapy for cardiac repair are
prove angiogenesis, preserve gap junctions, and enhance cardiac reviewed in the following sections.
functions compared to the undifferentiated MSCs [51,67,68].
3. Nanobiomaterial-incorporated stem cell therapy for
cardiac repair
2.2. Paracrine factor secretion of MSCs for cardiac repair
Mounting evidence for stem cell–derived paracrine action and stem
Even though the cardiac function improvements after MSC implan- cells’ cardiac lineage differentiation for cardiac repair has introduced a
tation were accompanied with the differentiation of MSCs, a previous crucial step forward for the development of biomaterial-integrated
study addressed that the number of newly differentiated or generated stem cell therapies. Recently, studies utilizing the nanoscale properties
cardiomyocytes was too small to explain the benefits of MSC therapy of biomaterials to potentiate the therapeutic efficacy of stem cells
[70]. After MI, more than a billion cardiomyocytes undergo apoptosis have made significant advances (Table 1). Biocompatible nanomaterials
and necrosis; hence, the small fraction of MSC-differentiated can deliver therapeutic molecules for angiogenesis and stem cell re-
cardiomyocytes may not elicit any meaningful benefit for attenuating cruitment, both of which promote cardiac repair. Additionally, integra-
cardiac remodeling post-MI. Growing evidence now suggests that tion of nanobiomaterials with stem cells may exploit synergistic
growth factors and cytokines secreted from the implanted MSCs play effects for cardiac repair by overcoming the limitations of conventional
a critical role in promoting cardiac repair [56,60]. MSCs secrete a variety stem cell therapy. Nanoscale properties of nanobiomaterials, such as
of growth factors and cytokines that exert protective effects on dam- nanotopography, electrical conductivity or unique chemical characteris-
aged tissues and organs [57–59]. These paracrine molecules are in- tics, may modulate stem cells’ behaviors needed for effective MI repair,
volved in cardiac tissue regeneration, neovascularization, cardiac including their structural alignment, cardiac differentiation, and para-
contractility, cardiac metabolism, myocardial protection, and even im- crine secretion. Additionally, nanoscale properties can also be used to
mune responses for cardiac repair [71]. deliver therapeutic molecules or cells for better treatment of MI. In the
The therapeutic effects of paracrine molecules for cardiac repair following sections, we provide an overview of the incorporation of
were demonstrated in the studies where the administration of condi- nanobiomaterials with stem cells for improved cardiac repair after MI
tioned medium of stem cell cultures recapitulated the benefits of stem (Fig. 2).
cell implantation [27,70]. Gnecchi et al. demonstrated that the condi-
tioned medium from MSC cultures showed cytoprotective effects on 3.1. Therapeutic molecule delivery systems
rat cardiomyocytes in vitro and significantly reduced cardiac fibrosis
in vivo [27]. Furthermore, paracrine factors secreted by the implanted The delivery of therapeutic molecules, including genes and proteins,
MSCs protected adjacent cardiomyocytes from undergoing cell death, can improve cardiac repair after MI by promoting angiogenesis [34,76]
attenuated left ventricular remodeling in an ischemic heart, and in- or recruiting stem cells [77]. To maximize the therapeutic effect of
duced angiogenesis and vasculogenesis in vivo [28]. To enhance the these biomolecules, sustained and controlled release of the drug is cru-
paracrine secretion of MSCs and elicit better tissue regeneration, previ- cial. Therefore, previous studies utilized nanobiomaterials to prolong
ous studies introduced hypoxic culture conditions or MSC spheroids the expression of proteins for improved angiogenesis and recruitment
generating mild hypoxia on in these cells [51,72–75]. It was also demon- of stem cells [31,32,35,78]. Research showed that the delivery of
strated that hypoxia preconditioning of MSCs in vitro not only platelet-derived growth factor-BB (PDGF-BB) using self-assembling
18 J. Han et al. / Advanced Drug Delivery Reviews 95 (2015) 15–28

Table 1
Results of biomaterials and biomaterials-integrated stem cell therapy for MI in animals or in vitro studies.

Approach and materials Cells or Model Delivery Time Major observations (in vitro and in vivo)
biomolecules method point
(in vivo)

Protein or gene delivery materials


Chitosan-alginate nanoparticle [31] PlGF Rat Intramyocardial 8 weeks Heart function ↑, vascular density ↑, scar formation ↓,
injection inflammation ↓
Self-assembling peptide nanofiber [35] PDGF-BB Rat Intramyocardial 2 weeks CMC death ↓, systolic function ↑, infarct size ↓, contractility ↑
(AcN-RARADADARARADADA-CNH2) injection
Baculovirus nanocomplex [91] Angiopoeitin-1 Rat Intramyocardial 4 weeks Capillary density ↑, infarct size ↓, heart function ↑
with ASC injection

Cell delivery materials


Self-assembling peptide [37] CSC Mouse Intramyocardial 2 weeks Heart function ↑, fibrosis ↓, cardiac differentiation ↑
(AcN-RADARADARADARADA-CNH2) injection
Collagen patch [97] MSC Rat Epicardial 4 weeks MSC engraftment ↑, wall thickness ↑, heart function ↑
surface patch
PLCL patch [98] MSC Rat Epicardial 4 weeks Heart function ↑, MSC cardiac differentiation ↑, infarct size ↓,
surface patch
Self-assembling peptide nanofiber [101] MNC Pig Intramyocardial 4 weeks Heart function ↑, engraftment ↑, capillary density ↑
(AcN-RARADADARARADADA-NH2) injection
Self-assembling peptide nanofiber [103] MSC Rat Intramyocardial 4 weeks Heart function ↑, infarct size ↓, capillary density ↑,
(AcN-RARADADARARADADA-NH2) injection
Self-assembling peptide nanofiber [104] CSC Rat Intramyocardial 4 weeks Vascular enlargement ↑, heart function ↑, capillary density ↑
(AcN-RADARADARADARADA-CNH2) injection

Nanomaterials with topographical cues


Patterned PEG hydrogel [41] CSC Rat Epicardial 3 weeks Wall thickness ↑, organized collagen deposition ↑, surviving
surface patch muscle cells ↑,
Microelectrode arrays [69] (in vitro study) MSC and CMC N/A N/A N/A MSC conduction velocity ↑, MSC gap junction protein ↑
co-culture
PCL nanofiber [106] MSC Rat Epicardial 4 weeks Heart function ↑, fibrosis ↓, wall thickness ↑
surface patch

Nanomaterials with electrical conductivity


Carbon nanotube [47] (in vitro study) MSC N/A N/A N/A MSC cardiac-specific and gap junction protein ↑
Gold nanoparticle embedded BSA/PVA scaffold MSC with N/A N/A N/A MSC cardiac-specific and gap junction protein ↑
[111] (in vitro study) 5-azacytidine
Reduced graphene oxide [112] MSC spheroids Mouse Intramyocardial 2 weeks MSC gap junction protein ↑, heart function ↑
injection

Nanomaterials with chemical cues


Iron oxide nanoparticle [51] MSC and H9C2 Rat Intramyocardial 2 weeks MSC cardiac-specific and gap junction protein ↑, MSC paracrine ↑,
co-culture injection heart function ↑, capillary density ↑
Self-assembling peptide nanofiber [114] MSC-conditioned Mouse Intramyocardial 4 weeks Contractility ↑, vascularization ↑, heart function ↑
(Not specified) medium injection
Graphene [115] (in vitro study) MSC N/A N/A N/A MSC cardiac-specific and gap junction protein ↑
Graphene [121] (in vitro study) ESC N/A N/A N/A ESC cardiac differentiation ↑

Peptide sequence: R, arginine; A, alanine; D, aspartic acid.

peptide nanofibers [35] or placental growth factor (PlGF) using peptide carriers showed no improvements in cardiac function. Similar
chitosan-alginate nanoparticles [31] significantly attenuated cardiac re- to the self-assembling peptide nanofibers that prolonged the release
modeling and improved cardiac function after MI. PDGF-BB is a well- of therapeutic molecules, chitosan-alginate nanoparticles were also
known protein that promotes angiogenesis [79]; however, PDGF-BB used to deliver angiogenic proteins in a sustainable manner [31]. PlGF
injected in vivo is rapidly cleared from the blood [80] and results in is another protein that stimulates angiogenesis [81]. Similar to PDGF-
low therapeutic efficacy. To overcome this problem and to prevent BB, rapid clearance of PlGF after in vivo injection significantly impedes
rapid clearance of the growth factor in vivo, Hsieh et al. utilized self- the function of PlGF for therapeutic angiogenesis. Previously,
assembling peptides in protein delivery [35]. Self-assembling peptides Binsalamah et al. developed chitosan-alginate nanoparticles that could
are composed of short peptide sequences linked to a fatty acid tail, solve this problem, and showed over a 120-h period of sustained release
and can spontaneously assemble into nanofibers under various physio- of PlGF in vitro using these nanoparticles. When injected in vivo, PlGF-
logical conditions. These peptides are biodegradable and suitable for bound nanoparticles significantly improved cardiac function and pro-
functional modifications, making them a good candidate for the treat- moted vessel regeneration due to the sustained expression of PlGF at
ment of MI. The study showed that PDGF-BB delivered without self- the damaged region. Overall, these studies demonstrate that
assembling peptides rapidly disappeared from the injected site after nanobiomaterials, which provide growth factor protection from local
24 h, and only a negligible amount of this protein was detected after enzymatic degradation and sustained release of the protein at the injec-
3 days. Conversely, self-assembling peptide nanofibers loaded with tion site, can greatly enhance the therapeutic effects of drug delivery for
PDGF-BB achieved a sustained release of PDGF-BB at the injection sites MI repair.
for more than 14 days. This is mainly because PDGF-BB was tightly In addition to the direct delivery of therapeutic proteins to the MI re-
bound to the self-assembling peptide nanofibers, which served as a res- gion, nanobiomaterials have also been engaged in the delivery of genes.
ervoir for the slow release of PDGF-BB at the infarcted region. Conse- Viral vector-based gene delivery, a commonly used method for gene
quently, the prolonged release of PDGF-BB from self-assembling transfection may be immunogenic and oncogenic and thus its clinical
peptide nanofibers protected the cardiac cells in vivo and enhanced car- use is limited [82]. Therefore, non-viral nanomaterials have attracted
diac function after MI. In contrast, the delivery of PDGF-BB without significant attention to replace viral vectors for gene delivery [83].
J. Han et al. / Advanced Drug Delivery Reviews 95 (2015) 15–28 19

Fig. 2. Integration of nanobiomaterials with stem cells for the treatment of MI. Nanobiomaterials can enhance the delivery efficacy of therapeutic molecules and cells for cardiac repair. In
addition, the nanotopographical, electrical, and chemical cues of nanobiomaterials can regulate stem cell functions for enhanced therapeutic efficacy for the treatment of MI.

Nanocarriers could prevent genes from endonuclease degradation, in- nanobeads (MNBs) were integrated with vectors. The conjugation of
hibit unwanted interactions with proteins, protect genes from immune MNBs with adenoviral vectors (MNBs/Ad) enabled higher gene trans-
detection, and prolong gene circulation time for target delivery [83]. Che duction efficiency in cultured cells under magnetic field stimulation
et al. utilized polycaprolactone (PCL)/polyethylenimine (PEI) nanofi- compared to Ad alone in vitro. Furthermore, the epicardial placement
bers to deliver disulfide bond–cross-linked branched PEI nanoparticles of a magnet effectively attracted the MNBs/Ad complexes to the injured
complexed with vascular endothelial growth factor (VEGF) genes myocardium when complexes were intravenously injected in vivo.
(ssPEI/VEGF nanoparticles) to cells cultured on nanofibers [32]. The Overall, this gene delivery approach using MNBs significantly enhanced
pores between PCL/PEI nanofibers enabled cells to grow in three dimen- the gene transfection efficiency in vivo, and reduced cardiac remodeling
sions, and provided adequate mechanical properties for improved cell for improved left ventricular function [78].
adhesion and proliferation. Moreover, non-specific adsorption of For stem cell homing and tissue repair, the delivery of stromal cell-
ssPEI/VEGF nanoparticles occurred uniformly over the surface of the derived factor 1 (SDF-1) showed significant potential [85,86]. SDF-1 is
nanofibers, resulting in increased transgene uptake by the cultured a chemokine involved in stem cell homing [87]. The expression of
cells. The nanofibers had a high surface-to-volume ratio making it pos- SDF-1 is stimulated by the induction of MI, which in turn, recruits
sible to immobilize a large quantity of ssPEI/VEGF nanoparticles. In ad- stem cells to the injured myocardium [86]. The migration of stem cells
dition, direct contact between the nanofiber-immobilized ssPEI/VEGF to the infarcted myocardium was further enhanced with SDF-1 over-
nanoparticles and cells significantly reduced the amount of genes re- expression by SDF-1 gene delivery to the left ventricular wall using an
quired for transfection compared to the bolus delivery of the nanoparti- adenovirus vector [86]. However, locally delivered SDF-1 protein is vul-
cles. Compared to branched PEI, a conventional polycation polymer nerable to rapid diffusion and cleavage. To overcome this problem,
used for gene transfection [84], the use of ssPEI to form nanoparticles Segers et al. designed SDF-1 that is resistant to cleavage, and tethered
with VEGF significantly reduced cytotoxicity and showed better trans- this chemokine to self-assembling peptides (Fig. 3) [88]. The chemotac-
fection efficiency of VEGF. Even though the utilization of PCL/PEI nano- tic activity of SDF-1 was better preserved when SDF-1 was locally deliv-
fibers has not been tested in vivo, this study showed that the ered with self-assembling peptides, and strong tethering of SDF-1 to
combination of nanofibers or nanoparticles with conventional gene de- self-assembling peptide nanofibers showed controlled release of the
livery methods could suggest a promising gene therapy approach for chemokine. SDF-1 protein was detectable for at least 7 days after injec-
treating MI [32]. tion only when it was delivered together with self-assembling peptide
In addition to nanofibers, Zhang et al. introduced magnetic nanofibers. Subsequently, delivery of nanofiber-integrated SDF-1 signif-
nanobeads for enhanced gene delivery efficacy of VEGF-encoded adeno- icantly enhanced stem cell homing, increased capillary density, and im-
viral vectors (AdhVEGF) for the treatment of MI [78]. One of the major proved cardiac function [88].
drawbacks of the systemic infusion of vectors to treat MI is the low Nanobiomaterials can also be used to transfect cells in vitro prior to
targeting efficacy at the infarct region. Liver cells mostly take up the sys- implantation. For example, various types of nanobiomaterials effective-
temically delivered vectors, and only a fraction of infused vectors are ly transfected genes into cardiac progenitor cells [89], skeletal myo-
targeted to the MI region. To overcome this limitation and promote ac- blasts [90], or adipose-derived stem cells [91]. Transfection of insulin
tive targeting of adenoviral vectors to the infarcted region, magnetic growth factor-1 (IGF-1) into cardiac progenitor cells using self-
20 J. Han et al. / Advanced Drug Delivery Reviews 95 (2015) 15–28

Fig. 3. Schematic illustration of nanobiomaterials utilized as vehicles for therapeutic molecule delivery for the treatment of MI. Incorporation of stem cell–recruiting SDF-1 fusion proteins
in self-assembling peptides. SDF-1–bound self-assembling peptides form nanofibers in vivo, retain chemotactic activity of SDF-1, and exhibit local release of SDF-1 at the infarcted region
for prolonged therapy for cardiac repair. Reproduced with permission from Ref. [88].

assembling peptide nanofibers prior to implantation enhanced cell sur- reparative action of the implanted cells is critical to improve the out-
vival in the infarcted heart and promoted heart repair [89]. When PEI comes of stem cell therapy [93]. A number of nanobiomaterials have
was used to transfect VEGF genes into skeletal myoblasts, implantation been utilized as cell delivery systems, including cardiac patches and in-
of these transfected cells into the infarcted myocardium showed better jectable nanomaterials.
cell survival at the infarcted region and significantly improved cardiac Nanostructured cardiac patches and injectable nanomaterials were
repair compared to naïve skeletal myoblasts [90]. In addition, transfec- studied to improve stem cell retention after implantation in vivo [29,
tion of the angiopoietin-1 gene into adipose-derived stem cells using 94–96]. These cardiac patches are placed on the epicardial surface of
baculovirus nanocomplex prior to implantation promoted angiogenesis the infarcted heart and they require appropriate nanostructural and
in the infarcted heart with enhanced secretion of angiopoietin-1 [91]. chemical properties that are compatible with the native cardiac tissue.
Overall, these studies show that nanobiomaterials promote efficient For this reason, the fabrication of cardiac patches with collagen, the pre-
gene transfection of the cells, and propose a novel approach for cardiac dominant extracellular matrix protein in the heart tissue, has attracted
tissue engineering. significant attention due to its environmental compatibility [29]. Colla-
gen has a nanofibrillar structure and can bundle into a three-
3.2. Cell delivery vehicles dimensional scaffold such as cardiac patch, which mimics native cardiac
extracellular matrix. Simpson et al. previously demonstrated that colla-
Although stem cell therapy has emerged as a promising treatment gen patches seeded with MSCs improved MSC survival in vitro and re-
option for MI, its therapeutic benefit still remains limited due to poor stored cardiac function after implantation (Fig. 4) [97]. Compared to
engraftment and survival of the implanted cells [92]. The efficient deliv- previous studies that showed less than 11% of cell engraftment after im-
ery and prolonged survival of stem cells have posed major challenges plantation, MSCs delivered with collagen patches achieved a cell en-
for its application to treat MI [8]. Therefore, the development of new graftment of 23% and resulted in improved cardiac function. After
therapeutic strategies that enhance the survival and extend the injection, the survival and engraftment of MSCs is often limited by the

Fig. 4. Schematic illustration of nanobiomaterials utilized as vehicles for cell delivery for the treatment of MI. Stem cell–loaded collagen patch is placed on the epicardial surface of the
infarcted myocardial region and held with fibrin sealant. Stem cells implanted with collagen patch show significantly improved initial cell engraftment for better cardiac function improve-
ment compared to direct cell injection. Reproduced with permission from Ref. [97].
J. Han et al. / Advanced Drug Delivery Reviews 95 (2015) 15–28 21

inflammatory microenvironment at the infarct region and the leakage for the treatment of MI, researchers predominantly focused on the in-
of the implanted cells from the injection site. Collagen-based cardiac nate anisotropic property of the heart because cardiac-mimetic struc-
patch provided cell-friendly extracellular matrix for better cell survival, tural nanotopography could pose significant effects on the cells [9,10,
and uniformly fixed MSCs in proximity to the infarct area for better en- 29]. The heart is a constantly working, dynamic, and a complex organ.
graftment. Cardiac patches without MSCs showed no improvement in For the functional activity of the heart, the cardiac cells that comprise
the cardiac function, suggesting that the major therapeutic effect was the heart muscles require a quick and efficient signal transfer. Therefore,
attributed to the implanted stem cells. the alignment of the cardiac cells and proper gap junction protein distri-
Along with the natural materials such as collagen, synthetic mate- bution are required for efficient cell signal transduction and synchro-
rials have also been investigated for cardiac patch development [29]. nous contraction of the heart [105]. To mimic the anisotropic property
Among the various types of synthetic polymers explored, poly(lactide- of the heart, various nanotopography-associated approaches using
co-ε-caprolactone) (PLCL) was suggested as a suitable candidate for car- nanobiomaterials were introduced to modulate cell behaviors, includ-
diac patches due to its elasticity and biodegradability [98]. PLCL is a ing cell alignment, differentiation and paracrine secretion, and then in-
highly elastic polymer, which can form nanofibrous scaffolds, and may tegrated with stem cells for cardiac repair.
serve as an excellent matrix for cardiac tissue engineering applications. A previous study demonstrated that contact guidance of the cells
When MSCs were seeded onto a PLCL-based cardiac patch and im- by aligned nanotopography or electrical field stimulation is crucial in
planted onto an infarcted heart, both MSC survival and cardiac function the orientation and elongation of cardiomyocytes [38]. Interestingly,
were significantly improved compared to the direct injection of MSCs this study showed that while the combination of nanotopography
[98]. This is because the cardiac patch served as a mechanical extracel- and electrical stimulation can promote the contractile property of
lular matrix for MSC survival and allowed MSCs to differentiate into cardiomyocytes, the topographical cue might serve as a stronger de-
functional cardiomyocytes. The study further demonstrated that PLCL terminant of cardiomyocyte alignment than electrical field stimula-
cardiac patch alone could not restore cardiac function, while MSC tion. Several studies previously demonstrated that aligned
alone could only partially restore heart function and reduce fibrosis nanofibers, which mimic the nanostructures of natural extracellular
[98]. However, contradictory results were also reported in other cardiac matrices of the heart tissue [39], improve the alignment [40] and
patch studies, where implantation of cardiac patch alone improved car- beating of the cardiomyocytes [42]. Cardiomyocytes cultured on
diac function [99,100]. Therefore, further studies are required to com- aligned nanofibrous electrospun patches showed a higher beating
prehensively investigate the efficacy of acellular patches and their frequency and amplitude compared to those cultured on randomly
impact on stem cell therapy for cardiac repair. oriented fibrous patches [40]. Upon implantation in vivo, the
Bio-inspired self-assembling peptide nanofibers can be utilized as a cardiomyocyte-seeded aligned cardiac patch improved cardiac func-
cell-delivery vehicle for the treatment of MI [101–103]. Due to their bio- tion, while the cardiomyocytes-seeded randomly oriented patch re-
compatibility and biodegradability, self-assembling peptides are uti- sulted in serious deterioration in the cardiac function [42]. This study
lized not only in drug delivery but also in cell delivery for cardiac indicates that the anisotropic property of nanobiomaterials signifi-
repair. Previous studies exploiting self-assembling peptides as cell- cantly affects the therapeutic outcomes of cell therapy for the treat-
delivery vehicles showed great improvement in the survival of the im- ment of MI, and is crucial for developing biomaterial-based therapy
planted cells and facilitate myocardial regeneration after MI for MI.
[101–103]. This is primarily because self-assembling peptides may in- The anisotropic properties of nanobiomaterials further affected the
crease cell retention and promote cell survival after in vivo delivery. To stem cell behavior for higher therapeutic efficacy. Kim et al. have
further elucidate the function of self-assembling peptides, Cui et al. previously shown that nanotopographically defined polyethylene glycol
demonstrated that these peptide nanofibers can enhance growth, sur- (PEG) hydrogel, which mimics the structural features of the native myo-
vival, and differentiation of MSCs in vitro [103]. These peptides sponta- cardial matrix can augment adhesion, migration, and proliferation of
neously assembled into nanofiber scaffolds that mimic the natural CSCs [41]. Implantation of stem cell–seeded anisotropic PEG scaffolds
extracellular matrix post-implantation. Consequently, MSCs mixed showed successful integration with the host tissue due to the
with self-assembling peptides exhibited increased cell survival and re- cardiac extracellular matrix-mimicking nanostructures. Moreover,
tention at the infarct size in vivo, resulting in improved cardiac function nanopatterned PEG scaffold significantly improved retention and
[103]. Furthermore, Guo et al. integrated self-assembling peptides with growth of transplanted CSCs. Consequently, the implantation of CSC-
cardiac stem cells and demonstrated their therapeutic application in rat seeded aligned cardiac patch greatly reduced cardiac fibrosis and
MI model [104]. To enhance survival and cardiac differentiation of CSCs improved cardiac function [41]. The anisotropic properties of
in vivo, Guo et al. constructed self-assembling peptides tethered with nanobiomaterials also improved the paracrine action of stem cells.
the cell-adhesion motif RGDSP. When these peptides were implanted Kang et al. recently demonstrated that MSCs cultured on aligned
together with CSCs, they bundled into nanofiber scaffolds, providing a fibronectin-immobilized PCL nanofibers exhibited enhanced expression
suitable microenvironment for CSC adhesion and survival, and of angiogenic and cardioprotective genes [106]. This study showed that
protected CSCs from apoptosis and necrosis caused by anoxia. Increased MSCs cultured on anisotropic fibronectin-immobilized PCL nanofibers
survival of the CSCs further enhanced their cardiac differentiation and exhibited aligned cellular phenotypes found in native cardiac tissue.
resulted in better cardiac repair. Compared to the groups treated with Furthermore, fibronectin immobilization significantly elevated the ex-
either CSCs or self-assembling peptides alone, the combination of CSCs pression of genes involved in stem cell homing, anti-apoptosis, anti-
and peptides significantly improved cardiac function and reduced colla- inflammation, and angiogenesis in MSCs. Subsequently, implantation
gen deposition. Collectively, these studies suggest that self-assembling of MSC-seeded anisotropic scaffolds significantly improved the cardiac
peptides can serve as an efficient cell delivery vehicle increasing cell function in rat MI model (Fig. 5) [106]. The mechanisms for the
survival, retention, and differentiation, and thereby promoting the ther- fibronectin-immobilized PCL nanofiber-mediated cellular signaling
apeutic efficacy of stem cell therapy for the treatment of MI. and paracrine molecule expression were not clarified in this study.
Thus, further studies differentiating the effects of fibronectin immobili-
3.3. Nanotopographical cues of nanobiomaterials zation from nanofiber alignment are required. Overall, even though the
above-mentioned studies by Kim et al. [41] or Kang et al. [106] did not
Nanobiomaterials with nanotopographical cues have been applied elucidate the mechanisms involved in cell behavior modulation on
in cell therapy to modulate cell behaviors such as cell differentiation, nanotopographically designed polymer scaffold, they certainly show
gap junction protein expression, and paracrine molecule secretion for that developing nanobiomaterials mimicking the nanostructures of car-
cardiac tissue repair. For development of therapeutic nanobiomaterials diac extracellular matrix is required for improved cardiac repair.
22 J. Han et al. / Advanced Drug Delivery Reviews 95 (2015) 15–28

Fig. 5. Schematic representation of nanotopography of nanobiomaterials utilized for cardiac tissue engineering. Poly(glycidyl methacrylate) (pGMA)-coated aligned polycaprolactone
(PCL) nanofibers were prepared, and fibronectins were immobilized onto the pGMA-PCL nanofibers. MSCs seeded on the nanofibers exhibit aligned cellular phenotypes found in natural
extracellular matrix of the heart. The MSCs-seeded fibronectin-immobilized aligned nanofibrous cardiac patches implanted onto a rat MI model show better tissue compatibility and sig-
nificantly improved cardiac function. Reproduced with permission from Ref. [106].

To further analyze the effect of cell alignment on the intercellular in- researchers have incorporated these nanomaterials with cell therapy
tegration and electrical conduction of stem cells, a previous study intro- for treating MI.
duced cellular alignments during MSC-cardiomyocyte co-culture [69]. A number of previous studies showed that integration of carbon
Pijnappels et al. showed that a forced alignment of MSCs on a nanofibers or carbon nanotubes with polymer scaffolds promotes the
directionally patterned substrate significantly improved cardiac differ- adhesion and proliferation of cardiomyocytes [43] and cardiac progeni-
entiation of MSCs and their functional integration with cardiomyocytes tor cells [44]. In addition, these conductive nanomaterials significantly
in the MSC-cardiomyocyte co-culture [69]. This study further demon- promoted the expression of Cx43 in cardiac cells and stimulated the
strated the alignment-dependent increase in MSCs’ conduction velocity electrical coupling between the cells. Cx43 expression is particularly im-
and expression of the gap junction protein, connexin 43 (Cx43). This portant for MI repair because it is essential for intercellular coupling, ar-
study shows that nanopatterns not only regulate the behaviors of the rhythmic risk reduction, and functional cardiomyogenic differentiation
cells cultured on scaffolds but also affect the synchronous integration of the cells [67,73,105,110]. Although the mechanisms for Cx43 upregu-
of MSCs with the surrounding cardiac environment. Taken together, lation have not been clearly elucidated, the overexpression of Cx43 in
the aforementioned studies suggest that cell alignment contributes to cells cultured on conductive scaffold suggests a potential use of conduc-
the therapeutic outcomes of cell therapy, and the development of tive nanobiomaterials for cardiac repair. To better understand the ther-
nanobiomaterials with proper anisotropic property is desirable for suc- apeutic potential of conductive materials and analyze the beating
cessful cardiac repair. behavior of the cells on a conductive scaffold, Martines et al. and
Kharaziha et al. investigated the electrophysiological function of
3.4. Electrically conductive nanobiomaterials cardiomyocytes seeded on a chitosan matrix incorporated with carbon
nanofibers [48] or poly(glycerol sebacate):gelatin scaffolds embedded
Along with the topographical property, electrical conductivity is an- with carbon nanotubes [50], respectively. Cardiac cells seeded on a con-
other unique feature of the heart that is critical in developing therapeu- ductive nanobiomaterial-integrated scaffold exhibited a synchronous
tic biomaterials for cardiac repair. Unlike cells in other organs, cardiac beating behavior and an increased expression of cardiac-specific genes
cells need gap junction–based intercellular coupling and electrical con- involved in cardiac muscle contraction and electrical coupling. Taken to-
duction for natural heart function. Thus, electrochemically unsuitable gether, the aforementioned studies demonstrate that conductive
therapies may pose electrophysiological discordance with native cardi- nanobiomaterials not only enhance the electrical coupling between
ac tissues, and may cause arrhythmias upon application [105]. There- the cells with Cx43 upregulation, but also induce synchronous beating
fore, it has been speculated that electrical conductivity may modulate of the cardiac tissue. The in vivo application of carbon nanomaterial–in-
the behaviors of stem cells and affect their electrochemical signaling tegrated scaffolds and the mechanisms involved in how the conductive
pathways that contribute to cardiac tissue integration. To mimic and materials affect cell behaviors require further studies.
preserve the electrical conductivity of the heart, previous studies fo- Gold nanomaterials were also utilized to improve the therapeutic ef-
cused on the development of conductive nanobiomaterial-integrated fects of cell therapy. Cardiomyocytes cultured in hydrogel scaffolds inte-
therapeutics that are electrically compatible with native cardiac grated with gold nanoparticles [45] or gold nanowires [46] showed an
tissues. Various nanobiomaterials, including carbon- and gold-based increased Cx43 expression compared to cardiomyocytes cultured in
nanomaterials, showed electrical conductivity [107–109], and hydrogels without the integration of gold nanomaterials. Dvir et al.
J. Han et al. / Advanced Drug Delivery Reviews 95 (2015) 15–28 23

Fig. 6. Schematic representation of electrically conductive nanobiomaterials utilized for cardiac tissue engineering. Incorporation of electrically conductive RGO flakes in MSC spheroids
greatly improves the expression of the gap junction protein Cx43, which is critical in native cardiac tissue compatibility and function. RGO-incorporated MSC spheroids are directly injected
into the infarcted region of a rat heart for greater cardiac function improvements compared to MSC spheroids incorporated with insulating graphene oxide flakes. Reproduced with per-
mission from Ref. [112].

incorporated gold nanowires into porous alginate scaffolds to develop Conductive nanobiomaterials have further been utilized together
three-dimensional conductive nanocomposites [46]. Non-conducting with conventional cardiac differentiation methods to stimulate car-
alginate pore walls bridged with conductive gold nanowires demon- diac differentiation of MSCs. Ravichandran et al. demonstrated that
strated enhanced electrical signal propagation, and enabled electrical 5-azacytidine treatment on MSCs seeded onto gold nanoparticle–
communication between the cardiomyocytes seeded on the scaffolds. embedded conductive Bovine serum albumin (BSA)/Polyvinyl alco-
Consequently, cardiomyocytes grown on the conductive nanocompos- hol (PVA) nanofibrous scaffolds greatly stimulated the MSC differen-
ite showed a better aligned tissue structure, and exhibited higher levels tiation towards cardiomyogenic lineage and enhanced Cx43
of the proteins involved in muscle contraction. The cells demonstrated expression [111]. This study indicates that the electrical conductivity
synchronous contraction when the conductive nanocomposites were of nanobiomaterials can exert synergistic effects with conventional
electrically stimulated [46]. Similar to the studies with carbon-based cardiac differentiation methods and that the combined approach
nanomaterials, this study suggests that the integration of gold can be a superior therapy for MI. Park et al. further demonstrated
nanomaterials with polymeric scaffolds may improve the therapeutic the therapeutic effect of conductive nanomaterials in vivo, and re-
efficacy of cell therapy. cently showed that the conductive and intrinsic chemical properties
To determine whether conductive nanomaterials can provide of reduced graphene oxide (RGO) can have synergistic effects to
cardiomimetic cues to MSCs for cardiac differentiation, a number of modulate the MSC function for the treatment of MI (Fig. 6) [112].
studies investigated the MSCs’ phenotype changes upon treatment RGO is composed mostly of carbon atoms and it has unique mechan-
with conductive materials. Mooney et al. demonstrated that electri- ical, chemical, and electrical properties [113]. The study showed that
cal stimulation of carbon nanotube–embedded scaffolds significantly the incorporation of RGO flakes in MSC spheroids significantly en-
enhanced the cardiomyogenic differentiation of MSCs seeded on the hanced the Cx43 expression of MSCs. In contrast, graphene oxide
scaffolds [47]. Electrical stimulation reoriented the cells perpendicu- flakes, which are insulating due to the existence of functional groups
lar to the direction of the current and significantly augmented the that hinder the transfer of π-electrons, significantly reduced the
expression of cardiac-specific biomarkers including Cx43. Without Cx43 expression in MSCs. Based on this data, the authors postulated
the carbon nanotubes, however, electrical stimulation alone did not that the enhanced expression of Cx43 in MSCs is attributed to the
promote cardiac protein expression in MSCs. To further differentiate electrical conductivity and cell-extracellular matrix interactions
the effect of electrical stimulation from the conductive materials, generated by RGO flakes. Ultimately, the implantation of MSC spher-
Crowder et al. further investigated carbon nanotube-containing syn- oids integrated with RGO flakes significantly reduced tissue fibrosis
thetic polymer scaffolds without electrical stimulation on the cardiac and improved cardiac function.
differentiation of MSCs [49]. They observed that the electrically con- Conductive nanobiomaterials have actively participated in intercel-
ductive nanobiomaterials alone promoted cardiac differentiation of lular coupling and cardiac differentiation, showing great potential in
the stem cells and their Cx43 expression without any electrical stim- therapeutic applications for the treatment of MI. However, the mecha-
ulation. These studies, however, did not clarify the mechanisms in- nisms behind their effects on stem cell behaviors still remain elusive.
volved in cardiac differentiation and Cx43 enhancement of MSCs Moreover, conductive nanobiomaterials need to properly mimic the
cultured on conductive materials. Further studies elucidating the ef- electrical conductivity of the native heart to reduce the arrhythmic
fects of conductive materials on cell behavior modulation and differ- risks of in vivo application [105]. Future studies elucidating the
entiation are therefore required for the therapeutic application of mechanisms involved in MSC behavior modulation by conductive
conductive materials for MI. nanomaterials, and the development of in vivo mimetic conductive
24 J. Han et al. / Advanced Drug Delivery Reviews 95 (2015) 15–28

materials would provide a better therapeutic window for the applica- H9C2 cells [120] limits the effectiveness of MSC cardiac priming by
tion of conductive nanobiomaterials for cardiac repair. MSC-H9C2 co-culture [51]. Han et al. recently showed that the inter-
nalization of iron oxide nanoparticles can augment the Cx43 expres-
3.5. Intrinsic chemistry of nanobiomaterials sion in H9C2 cells by delivering metal ions and modulating the
intracellular c-Jun N-terminal kinase signaling [51]. Changes in in-
Nanobiomaterials with distinct chemical properties can also be ap- tracellular ion levels can induce significant alteration in cell signal-
plied to stem cell therapy for MI. Previous studies speculated that nano- ing, and may cause cell cytotoxicity. Hence, Han et al. speculated
scale properties of nanobiomaterials might pose unique chemical that a change in the concentration of intracellular iron ion level
characteristics that can affect adhesion, growth, differentiation and might change cell signaling pathways. Conforming to previous stud-
paracrine secretion of stem cells. Nanobiomaterials with functional ies, exogenous delivery of metal ions caused significant toxicity to
chemical properties can capture growth factors secreted by stem cells the cells. However, using iron oxide nanoparticles, Han et al. success-
and be used for acellular therapies for MI [114], stimulate stem cells to fully increased metal ion levels within the cells without cytotoxicity,
secrete more paracrine molecules for enhanced therapeutic efficacy and initiated cell signaling alteration for Cx43 upregulation. The en-
[112], induce differentiation of stem cells toward cardiac lineage [115], hanced Cx43 expression in the H9C2 cells further stimulated the in-
and control intracellular signaling pathways for cell function tercellular crosstalk between the H9C2 cells and MSCs. Hence, MSCs
modulation [51]. co-cultured with iron oxide nanoparticle–internalized H9C2 cells
Self-assembling peptide nanofibers that capture paracrine mole- showed improved cardiac differentiation and paracrine molecule se-
cules have been utilized for MI repair [114]. Webber et al. demon- cretion. Consequently, the implantation of co-cultured MSCs re-
strated that heparin-presenting self-assembling peptides can duced fibrosis, enhanced angiogenesis, and improved cardiac
bundle into a matrix consisting of fibrillar nanostructures similar to function (Fig. 7A) [51].
the natural cardiac extracellular matrix [114]. The presentation of In addition to iron oxide nanoparticles, carbon-based
heparin on the surface of self-assembling peptide nanofibers further nanomaterials have also shown significant potential in stimulating
enabled the binding of growth factors secreted from the stem cells in cell signaling and cardiac differentiation of stem cells. Park et al. re-
cell cultures via heparin-binding domains [114]. Using recombinant cently showed that graphene, a single layer of carbon atoms stimu-
proteins of VEGF and basic fibroblast growth factor (bFGF) loaded lated the MSC differentiation process toward cardiomyogenic
in the nanofibers, the effects of the nanofiber-captured growth fac- lineage [115]. When MSCs were cultured on graphene, its unique
tors from the stem cell–conditioned medium were recapitulated. chemical nanostructure stimulated the expression of the extracellu-
When heparin-presenting self-assembling peptides were incubated lar matrix and signaling molecules related to cardiomyogenic differ-
with hypoxic stem cell conditioned medium, they captured more entiation, and in turn, enhanced cardiomyogenic gene expression in
than 58% of bFGF present in the medium. When implanted into the MSCs (Fig. 7B) [115]. Lee et al. also demonstrated that graphene pro-
damaged heart, the growth factor–bound nanofibers successfully re- motes the cardiac differentiation of ESCs [121]. ESCs cultured on
stored the hemodynamic function of the heart by modulating the graphene exhibited significant changes in the extracellular signal-
cardiac metabolism [114]. This study shows the therapeutic poten- regulated kinase and the focal adhesion kinase signaling compared
tial of synthetic nanomaterials in combination with stem cell para- to ESCs cultured on glass or matrigel. Consequently, ESCs cultured
crine factors for the treatment of MI. on graphene showed great increase in the expression of genes asso-
Cell-sheet engineering also serves as a treatment option for the ciated with cardiac transcription, early cardiomyocytes, late
treatment of MI. Implantation of stem cell sheets can significantly cardiomyocytes, and gap junctions [121].
enhance the stem cells’ capability to repair the injured myocardium The intrinsic chemistry of nanobiomaterials also contributed to the
compared to the implantation of dissociated stem cells [116]. enhanced paracrine secretion and Cx43 expression of MSCs cultured
Okura et al. have previously demonstrated successful treatment of in spheroid forms (Fig. 6) [112]. MSCs in spheroid forms have very lim-
MI using MSCs cultured on thermo-responsive dishes [117]. The ited cell-extracellular matrix interactions, which are necessary for para-
study demonstrated that the implantation of cell sheets composed crine secretion and cell survival. Recently, Park et al. demonstrated that
of MSC-derived cardiomyoblast-like cells resulted in functional dif- RGO efficiently adsorbs extracellular matrix proteins via π-π stacking
ferentiation of the cells towards cardiomyocytes in vivo and en- and hydrophobic interactions [122], providing better cell-extracellular
hanced cardiac function [117]. With the unique thermo-responsive matrix interactions for MSCs in spheroids [112]. When RGO flakes
properties of poly(N-isopropylacrylamide) (PIPAAm), PIPAAm- were cultured in serum-containing cell culture medium, significant ad-
grafted surface provides a useful tool to form cell sheets as it allows sorption of extracellular matrix protein fibronectin was observed on the
cells to adhere and detach depending on the temperature [116]. Fur- surface of RGO flakes. Furthermore, incorporation of these RGO flakes in
ther studies demonstrated that nanoscale thickness of PIPAAm coat- MSC spheroids increased the expression of cell signaling molecules
ing on the surface plays a crucial role in the formation and associated with cell-extracellular matrix interactions. The enhanced
detachment of the cell-sheets [118,119]. Akiyama et al. showed cell-extracellular matrix interactions then stimulated paracrine mole-
that cell adhesion on PIPAAm-grafted surface is inhibited when the cule secretion and Cx43 expression. The implantation of these RGO
coating thickness of PIPAAm increases from 15.5 ± 7.2 nm to flake-incorporated MSC spheroids significantly improved the cardiac
29.5 ± 8.4 nm, because PIPAAM grafting inhibits adsorption of cell function in mouse MI models compared to MSC spheroids [112]. Collec-
adhesion proteins like fibronectin to the surface [118]. tively, previous studies showed that the integration of nanobiomaterials
Iron oxide nanoparticles have also been utilized for cell function with stem cells improves the therapeutic outcomes of conventional
modulation and cardiac repair due to their innate chemistry. Widely stem cell therapy for the treatment of MI. Hence, defining the intrinsic
used for in vivo imaging, iron oxide nanoparticles were recently ap- chemical properties of the nanomaterials that can influence the stem
plied to modify the cell functions of H9C2 cells, a cardiomyoblast cell function would provide a basis for the development of better thera-
cell line, in an MSC-H9C2 co-culture [51]. The co-culture of MSCs peutic options for future MI therapies.
with cardiac cells previously demonstrated priming of MSCs toward
cardiac lineage and the implantation of these cardiac primed-MSCs 4. Conclusions
into the infarcted heart improved cardiac function [67]. For co-
culture, the rat cardiomyoblast cell line H9C2 has attracted much Stem cell therapy has become one of the most encouraging treat-
attention due to its easy accessibility compared to primary ment options for MI. The simple delivery of stem cells has been applied
cardiomyocytes. However, the low level of Cx43 expression in in a number of preclinical and clinical studies; however, the therapeutic
J. Han et al. / Advanced Drug Delivery Reviews 95 (2015) 15–28 25

Fig. 7. Effects of intrinsic chemical properties of nanobiomaterials on cell function and cardiac repair. (A) Mechanisms involved in iron oxide nanoparticle–induced Cx43 expression en-
hancement in cardiomyoblast H9C2 cell line, and in vivo therapeutic efficacy of the co-cultured MSCs. *p b 0.05. Internalization of iron oxide nanoparticles affects intracellular JNK signaling
pathway for Cx43 upregulation, which is crucial in intercellular crosstalk between MSCs and H9C2. MSCs co-cultured with iron oxide nanoparticle–internalized H9C2 (cMSC) show better
cardiac lineage development and in vivo therapeutic efficacy. (B) Schematic diagram of the signal transduction pathways associated with cardiomyogenic differentiation and cell survival,
and Western blot analyses regarding the molecules involved in the signal transduction pathways of MSCs cultured on graphene or coverslips for 3 weeks. Graphene stimulates the expres-
sion of extracellular matrix and signaling molecules involved in cardiomyogenic differentiation of MSCs. Quantification of the relative protein expressions of the cell signaling molecules.
*p b 0.05 compared to MSCs cultured on coverslips. Reproduced with permission from Refs. [51,115].

efficacy of stem cells still needs improvement and the efficient clinical References
translation of stem cell therapy remains elusive. To improve the thera-
[1] M. Mirotsou, T.M. Jayawardena, J. Schmeckpeper, M. Gnecchi, V.J. Dzau, Paracrine
peutic efficacy of stem cells, nanobiomaterials have been utilized in a mechanisms of stem cell reparative and regenerative actions in the heart, J. Mol.
number of different forms. Nanobiomaterial-integrated strategies for Cell. Cardiol. 50 (2011) 280–289.
the treatment of MI include the delivery of therapeutic molecules or [2] D. Lloyd-Jones, R.J. Adams, T.M. Brown, M. Carnethon, S. Dai, G. De Simone,
T.B. Ferguson, E. Ford, et al., Heart disease and stroke statistics-2010 update:
cells, the development of cardiac niche-mimicking biomaterials with a report from the American Heart Association, Circulation 121 (2010)
electrical conductivity or anisotropic nanotopography, and the exploita- e46–e215.
tion of the chemical properties of nanomaterials. Nanoparticles and self- [3] H.J. Park, F. Yang, S.W. Cho, Nonviral delivery of genetic medicine for therapeutic
angiogenesis, Adv. Drug Deliv. Rev. 64 (2012) 40–52.
assembling peptides have been utilized in drug or cell delivery systems [4] A. Aguirre, I. Sancho-Martinez, J.C. Izpisua Belmonte, Reprogramming toward heart
to improve the delivery efficiency of the therapeutic molecules and or regeneration: stem cells and beyond, Cell Stem Cell 12 (2013) 275–284.
the cells at the infarcted myocardium. Additionally, nanotopographical [5] J.J. McMurray, M.A. Pfeffer, Heart failure, Lancet 365 (2005) 1877–1889.
[6] E. Bramucci, A. Repetto, M. Ferrario, U. Canosi, E. Boschetti, N. Brambilla, M.
cues and electrical conductivity of nanomaterials have been incorporat-
Gnecchi, P.A. Merlini, et al., Effectiveness of adjunctive stent implantation follow-
ed in nanobiomaterial designs to mimic the native properties of the ing directional coronary atherectomy for treatment of left anterior descending
heart and promote cardiomyocyte-like functions in stem cells. Finally, ostial stenosis, Am. J. Cardiol. 90 (2002) 1074–1078.
the interactions between the inherent chemical properties of [7] M. Gnecchi, P. Danieli, E. Cervio, Mesenchymal stem cell therapy for heart disease,
Vasc. Pharmacol. 57 (2012) 48–55.
nanobiomaterials with the stem cells were shown to affect the thera- [8] C.L. Hastings, E.T. Roche, E. Ruiz-Hernandez, K. Schenke-Layland, C.J. Walsh,
peutic behaviors of these stem cells. Taken together, the integration of G.P. Duffy, Drug and cell delivery for cardiac regeneration, Adv. Drug Deliv.
nanobiomaterials with stem cells showed a synergistic effect for the Rev. 84 (2015) 85–106.
[9] S. Pascual-Gil, E. Garbayo, P. Diaz-Herraez, F. Prosper, M.J. Blanco-Prieto, Heart re-
treatment of MI. The development and utilization of functional generation after myocardial infarction using synthetic biomaterials, J. Control. Re-
nanobiomaterials that can achieve cooperative therapy with stem cells lease 203 (2015) 23–38.
would provide a promising strategy for cardiac repair after MI in the [10] L.A. Reis, L.L. Chiu, N. Feric, L. Fu, M. Radisic, Biomaterials in myocardial tissue en-
gineering, J. Tissue Eng. Regen. Med. (2014) http://dx.doi.org/10.1002/term.1944.
future. [11] H.V. Huikuri, K. Kervinen, M. Niemela, K. Ylitalo, M. Saily, P. Koistinen, E.R.
Savolainen, H. Ukkonen, et al., Effects of intracoronary injection of mononuclear
bone marrow cells on left ventricular function, arrhythmia risk profile, and reste-
nosis after thrombolytic therapy of acute myocardial infarction, Eur. Heart J. 29
Acknowledgments
(2008) 2723–2732.
[12] G.P. Meyer, K.C. Wollert, J. Lotz, J. Pirr, U. Rager, P. Lippolt, A. Hahn, S. Fichtner, et al.,
This research was supported by the National Research Foundation of Intracoronary bone marrow cell transfer after myocardial infarction: 5-year
follow-up from the randomized-controlled BOOST trial, Eur. Heart J. 30 (2009)
Korea (2014073757) and the Korea Health Industry Development Insti-
2978–2984.
tute (KHIDI), Ministry of Health and Welfare (HI14C3270, HI14C1550 [13] B. Assmus, A. Rolf, S. Erbs, A. Elsasser, W. Haberbosch, R. Hambrecht, H. Tillmanns, J.
and HI15C0498), Republic of Korea. Yu, et al., Clinical outcome 2 years after intracoronary administration of bone
26 J. Han et al. / Advanced Drug Delivery Reviews 95 (2015) 15–28

marrow-derived progenitor cells in acute myocardial infarction, Circ. Heart Fail. 3 [37] M. Tokunaga, M.L. Liu, T. Nagai, K. Iwanaga, K. Matsuura, T. Takahashi, M. Kanda, N.
(2010) 89–96. Kondo, et al., Implantation of cardiac progenitor cells using self-assembling peptide
[14] D.M. Leistner, U. Fischer-Rasokat, J. Honold, F.H. Seeger, V. Schachinger, R. improves cardiac function after myocardial infarction, J. Mol. Cell. Cardiol. 49
Lehmann, H. Martin, I. Burck, et al., Transplantation of progenitor cells and regen- (2010) 972–983.
eration enhancement in acute myocardial infarction (TOPCARE-AMI): final 5-year [38] H.T. Au, I. Cheng, M.F. Chowdhury, M. Radisic, Interactive effects of surface topog-
results suggest long-term safety and efficacy, Clin. Res. Cardiol. 100 (2011) raphy and pulsatile electrical field stimulation on orientation and elongation of fi-
925–934. broblasts and cardiomyocytes, Biomaterials 28 (2007) 4277–4293.
[15] D.M. Hodgson, A. Behfar, L.V. Zingman, G.C. Kane, C. Perez-Terzic, A.E. Alekseev, M. [39] X. Zong, H. Bien, C.Y. Chung, L. Yin, D. Fang, B.S. Hsiao, B. Chu, E. Entcheva,
Puceat, A. Terzic, Stable benefit of embryonic stem cell therapy in myocardial in- Electrospun fine-textured scaffolds for heart tissue constructs, Biomaterials 26
farction, Am. J. Physiol. Heart Circ. Physiol. 287 (2004) H471–479. (2005) 5330–5338.
[16] J.Y. Min, Y. Yang, K.L. Converso, L. Liu, Q. Huang, J.P. Morgan, Y.F. Xiao, Transplanta- [40] C.W. Hsiao, M.Y. Bai, Y. Chang, M.F. Chung, T.Y. Lee, C.T. Wu, B. Maiti, Z.X. Liao, et al.,
tion of embryonic stem cells improves cardiac function in postinfarcted rats, J. Electrical coupling of isolated cardiomyocyte clusters grown on aligned conductive
Appl. Physiol. 92 (2002) 288–296. nanofibrous meshes for their synchronized beating, Biomaterials 34 (2013)
[17] C.Q. Xie, J. Zhang, Y. Xiao, L. Zhang, Y. Mou, X. Liu, M. Akinbami, T. Cui, et al., Trans- 1063–1072.
plantation of human undifferentiated embryonic stem cells into a myocardial in- [41] D.H. Kim, R.R. Kshitiz, P. Smith, E.H. Kim, H.N. Ahn, E. Kim, K.Y. Marban, Suh, et al.,
farction rat model, Stem Cells Dev. 16 (2007) 25–29. Nanopatterned cardiac cell patches promote stem cell niche formation and myo-
[18] L. Ye, Y.H. Chang, Q. Xiong, P. Zhang, L. Zhang, P. Somasundaram, M. Lepley, C. cardial regeneration, Integr. Biol. 4 (2012) 1019–1033.
Swingen, et al., Cardiac repair in a porcine model of acute myocardial infarction [42] Y.D. Lin, M.C. Ko, S.T. Wu, S.F. Li, J.F. Hu, Y.J. Lai, H.I.C. Harn, I.C. Laio, et al., A
with human induced pluripotent stem cell-derived cardiovascular cells, Cell nanopatterned cell-seeded cardiac patch prevents electro-uncoupling and im-
Stem Cell 15 (2014) 750–761. proves the therapeutic efficacy of cardiac repair, Biomater. Sci. 2 (2014)
[19] C. Templin, R. Zweigerdt, K. Schwanke, R. Olmer, J.R. Ghadri, M.Y. Emmert, E. 567–580.
Muller, S.M. Kuest, et al., Transplantation and tracking of human-induced pluripo- [43] D.A. Stout, B. Basu, T.J. Webster, Poly(lactic-co-glycolic acid): carbon nanofiber
tent stem cells in a pig model of myocardial infarction: assessment of cell survival, composites for myocardial tissue engineering applications, Acta Biomater. 7
engraftment, and distribution by hybrid single photon emission computed tomog- (2011) 3101–3112.
raphy/computed tomography of sodium iodide symporter transgene expression, [44] A.M. Wickham, M.M. Islam, D. Mondal, J. Phopase, V. Sadhu, E. Tamas, N. Polisetti,
Circulation 126 (2012) 430–439. A. Richter-Dahlfors, et al., Polycaprolactone-thiophene-conjugated carbon nano-
[20] Q. Xiong, L. Ye, P. Zhang, M. Lepley, J. Tian, J. Li, L. Zhang, C. Swingen, et al., Func- tube meshes as scaffolds for cardiac progenitor cells, J. Biomed. Mater. Res. B
tional consequences of human induced pluripotent stem cell therapy: myocardial Appl. Biomater. 102 (2014) 1553–1561.
ATP turnover rate in the in vivo swine heart with postinfarction remodeling, Circu- [45] J.O. You, M. Rafat, G.J. Ye, D.T. Auguste, Nanoengineering the heart: conductive
lation 127 (2013) 997–1008. scaffolds enhance connexin 43 expression, Nano Lett. 11 (2011) 3643–3648.
[21] K.U. Hong, Y. Guo, Q.H. Li, P. Cao, T. Al-Maqtari, B.N. Vajravelu, J. Du, M.J. Book, et al., [46] T. Dvir, B.P. Timko, M.D. Brigham, S.R. Naik, S.S. Karajanagi, O. Levy, H. Jin, K.K.
c-kit + Cardiac stem cells alleviate post-myocardial infarction left ventricular dys- Parker, et al., Nanowired three-dimensional cardiac patches, Nat. Nanotechnol. 6
function despite poor engraftment and negligible retention in the recipient heart, (2011) 720–725.
PLoS One 9 (2014) e96725. [47] E. Mooney, J.N. Mackle, D.J. Blond, E. O'Cearbhaill, G. Shaw, W.J. Blau, F.P. Barry, V.
[22] A. Schaefer, C. Zwadlo, M. Fuchs, G.P. Meyer, P. Lippolt, K.C. Wollert, H. Drexler, Barron, et al., The electrical stimulation of carbon nanotubes to provide a
Long-term effects of intracoronary bone marrow cell transfer on diastolic function cardiomimetic cue to MSCs, Biomaterials 33 (2012) 6132–6139.
in patients after acute myocardial infarction: 5-year results from the randomized- [48] A.M. Martins, G. Eng, S.G. Caridade, J.F. Mano, R.L. Reis, G. Vunjak-Novakovic, Elec-
controlled BOOST trial—an echocardiographic study, Eur. J. Echocardiogr. 11 trically conductive chitosan/carbon scaffolds for cardiac tissue engineering,
(2010) 165–171. Biomacromolecules 15 (2014) 635–643.
[23] M. Kudo, Y. Wang, M.A. Wani, M. Xu, A. Ayub, M. Ashraf, Implantation of bone mar- [49] S.W. Crowder, Y. Liang, R. Rath, A.M. Park, S. Maltais, P.N. Pintauro, W. Hofmeister,
row stem cells reduces the infarction and fibrosis in ischemic mouse heart, J. Mol. C.C. Lim, et al., Poly(epsilon-caprolactone)-carbon nanotube composite scaffolds
Cell. Cardiol. 35 (2003) 1113–1119. for enhanced cardiac differentiation of human mesenchymal stem cells,
[24] D. Orlic, J. Kajstura, S. Chimenti, D.M. Bodine, A. Leri, P. Anversa, Bone marrow stem Nanomedicine 8 (2013) 1763–1776.
cells regenerate infarcted myocardium, Pediatr. Transplant. 7 (Suppl. 3) (2003) [50] M. Kharaziha, S.R. Shin, M. Nikkhah, S.N. Topkaya, N. Masoumi, N. Annabi, M.R.
86–88. Dokmeci, A. Khademhosseini, Tough and flexible CNT-polymeric hybrid scaffolds
[25] M.F. Pittenger, B.J. Martin, Mesenchymal stem cells and their potential as cardiac for engineering cardiac constructs, Biomaterials 35 (2014) 7346–7354.
therapeutics, Circ. Res. 95 (2004) 9–20. [51] J. Han, B. Kim, J.Y. Shin, S. Ryu, M. Noh, J. Woo, J.S. Park, Y. Lee, et al., Iron oxide
[26] X. Hu, S.P. Yu, J.L. Fraser, Z. Lu, M.E. Ogle, J.A. Wang, L. Wei, Transplantation of nanoparticle-mediated development of cellular gap junction crosstalk to improve
hypoxia-preconditioned mesenchymal stem cells improves infarcted heart func- mesenchymal stem cells' therapeutic efficacy for myocardial infarction, ACS
tion via enhanced survival of implanted cells and angiogenesis, J. Thorac. Nano 9 (2015) 2805–2819.
Cardiovasc. Surg. 135 (2008) 799–808. [52] J.M. Hare, J.E. Fishman, G. Gerstenblith, D.L. DiFede Velazquez, J.P. Zambrano, V.Y.
[27] M. Gnecchi, H. He, N. Noiseux, O.D. Liang, L. Zhang, F. Morello, H. Mu, L.G. Melo, Suncion, M. Tracy, E. Ghersin, et al., Comparison of allogeneic vs autologous bone
et al., Evidence supporting paracrine hypothesis for Akt-modified mesenchymal marrow-derived mesenchymal stem cells delivered by transendocardial injection
stem cell-mediated cardiac protection and functional improvement, FASEB J. 20 in patients with ischemic cardiomyopathy: the POSEIDON randomized trial,
(2006) 661–669. JAMA 308 (2012) 2369–2379.
[28] R. Uemura, M. Xu, N. Ahmad, M. Ashraf, Bone marrow stem cells prevent left ven- [53] J.H. Houtgraaf, W.K. den Dekker, B.M. van Dalen, T. Springeling, R. de Jong, R.J. van
tricular remodeling of ischemic heart through paracrine signaling, Circ. Res. 98 Geuns, M.L. Geleijnse, F. Fernandez-Aviles, et al., First experience in humans using
(2006) 1414–1421. adipose tissue-derived regenerative cells in the treatment of patients with ST-
[29] A.A. Rane, K.L. Christman, Biomaterials for the treatment of myocardial infarction: segment elevation myocardial infarction, J. Am. Coll. Cardiol. 59 (2012) 539–540.
a 5-year update, J. Am. Coll. Cardiol. 58 (2011) 2615–2629. [54] K.C. Wollert, H. Drexler, Clinical applications of stem cells for the heart, Circ. Res. 96
[30] J.L. Ungerleider, K.L. Christman, Concise review: injectable biomaterials for the (2005) 151–163.
treatment of myocardial infarction and peripheral artery disease: translational [55] M.A. Laflamme, C.E. Murry, Heart regeneration, Nature 473 (2011) 326–335.
challenges and progress, Stem Cells Transl. Med. 3 (2014) 1090–1099. [56] L. Zeng, Q. Hu, X. Wang, A. Mansoor, J. Lee, J. Feygin, G. Zhang, P. Suntharalingam,
[31] Z.M. Binsalamah, A. Paul, A.A. Khan, S. Prakash, D. Shum-Tim, Intramyocardial et al., Bioenergetic and functional consequences of bone marrow-derived
sustained delivery of placental growth factor using nanoparticles as a vehicle for multipotent progenitor cell transplantation in hearts with postinfarction left ven-
delivery in the rat infarct model, Int. J. Nanomedicine 6 (2011) 2667–2678. tricular remodeling, Circulation 115 (2007) 1866–1875.
[32] H.L. Che, M. Muthiah, Y. Ahn, S. Son, W.J. Kim, H. Seonwoo, J.H. Chung, C.S. Cho, [57] D.F. Anthony, P.G. Shiels, Exploiting paracrine mechanisms of tissue regeneration
et al., Biodegradable particulate delivery of vascular endothelial growth factor to repair damaged organs, Transplant. Res. 2 (2013) 10.
plasmid from polycaprolactone/polyethylenimine electrospun nanofibers for [58] P.R. Baraniak, T.C. McDevitt, Stem cell paracrine actions and tissue regeneration,
the treatment of myocardial infarction, J. Nanosci. Nanotechnol. 11 (2011) Regen. Med. 5 (2010) 121–143.
7073–7077. [59] M.Z. Ratajczak, M. Kucia, T. Jadczyk, N.J. Greco, W. Wojakowski, M. Tendera, J.
[33] M.E. Davis, P.C. Hsieh, T. Takahashi, Q. Song, S. Zhang, R.D. Kamm, A.J. Grodzinsky, Ratajczak, Pivotal role of paracrine effects in stem cell therapies in regenerative
P. Anversa, et al., Local myocardial insulin-like growth factor 1 (IGF-1) delivery medicine: can we translate stem cell-secreted paracrine factors and microvesicles
with biotinylated peptide nanofibers improves cell therapy for myocardial infarc- into better therapeutic strategies? Leukemia 26 (2012) 1166–1173.
tion, Proc. Natl. Acad. Sci. U. S. A. 103 (2006) 8155–8160. [60] L.C. Amado, A.P. Saliaris, K.H. Schuleri, M. St John, J.S. Xie, S. Cattaneo, D.J. Durand, T.
[34] X. Hao, E.A. Silva, A. Mansson-Broberg, K.H. Grinnemo, A.J. Siddiqui, G. Dellgren, E. Fitton, et al., Cardiac repair with intramyocardial injection of allogeneic mesenchy-
Wardell, L.A. Brodin, et al., Angiogenic effects of sequential release of VEGF-A165 mal stem cells after myocardial infarction, Proc. Natl. Acad. Sci. U. S. A. 102 (2005)
and PDGF-BB with alginate hydrogels after myocardial infarction, Cardiovasc. 11474–11479.
Res. 75 (2007) 178–185. [61] Y.S. Yoon, A. Wecker, L. Heyd, J.S. Park, T. Tkebuchava, K. Kusano, A. Hanley, H.
[35] P.C. Hsieh, M.E. Davis, J. Gannon, C. MacGillivray, R.T. Lee, Controlled delivery of Scadova, et al., Clonally expanded novel multipotent stem cells from human
PDGF-BB for myocardial protection using injectable self-assembling peptide nano- bone marrow regenerate myocardium after myocardial infarction, J. Clin. Invest.
fibers, J. Clin. Invest. 116 (2006) 237–248. 115 (2005) 326–338.
[36] T.P. Kraehenbuehl, P. Zammaretti, A.J. Van der Vlies, R.G. Schoenmakers, M.P. [62] J.M. Duran, C.A. Makarewich, T.E. Sharp, T. Starosta, F. Zhu, N.E. Hoffman, Y. Chiba,
Lutolf, M.E. Jaconi, J.A. Hubbell, Three-dimensional extracellular matrix-directed M. Madesh, et al., Bone-derived stem cells repair the heart after myocardial infarc-
cardioprogenitor differentiation: systematic modulation of a synthetic cell- tion through transdifferentiation and paracrine signaling mechanisms, Circ. Res.
responsive PEG-hydrogel, Biomaterials 29 (2008) 2757–2766. 113 (2013) 539–552.
J. Han et al. / Advanced Drug Delivery Reviews 95 (2015) 15–28 27

[63] N. Nishiyama, S. Miyoshi, N. Hida, T. Uyama, K. Okamoto, Y. Ikegami, K. Miyado, K. [90] L. Ye, H. Haider, R. Tan, W. Toh, P.K. Law, W. Tan, L. Su, W. Zhang, et al., Transplan-
Segawa, et al., The significant cardiomyogenic potential of human umbilical cord tation of nanoparticle transfected skeletal myoblasts overexpressing vascular en-
blood-derived mesenchymal stem cells in vitro, Stem Cells 25 (2007) 2017–2024. dothelial growth factor-165 for cardiac repair, Circulation 116 (2007) I113–120.
[64] P. Antonitsis, E. Ioannidou-Papagiannaki, A. Kaidoglou, C. Papakonstantinou, In [91] A. Paul, M. Nayan, A.A. Khan, D. Shum-Tim, S. Prakash, Angiopoietin-1-expressing
vitro cardiomyogenic differentiation of adult human bone marrow mesenchymal adipose stem cells genetically modified with baculovirus nanocomplex: investiga-
stem cells. The role of 5-azacytidine, Interact. Cardiovasc. Thorac. Surg. 6 (2007) tion in rat heart with acute infarction, Int. J. Nanomedicine 7 (2012) 663–682.
593–597. [92] J. Terrovitis, R. Lautamaki, M. Bonios, J. Fox, J.M. Engles, J. Yu, M.K. Leppo, M.G.
[65] W.R. Xu, X.R. Zhang, H. Qian, W. Zhu, X.C. Sun, J. Hu, H. Zhou, Y.C. Chen, Mesenchy- Pomper, et al., Noninvasive quantification and optimization of acute cell retention
mal stem cells from adult human bone marrow differentiate into a cardiomyocyte by in vivo positron emission tomography after intramyocardial cardiac-derived
phenotype in vitro, Exp. Biol. Med. 229 (2004) 623–631. stem cell delivery, J. Am. Coll. Cardiol. 54 (2009) 1619–1626.
[66] Y. Huang, X. Jia, K. Bai, X. Gong, Y. Fan, Effect of fluid shear stress on [93] W.Y. Lee, H.J. Wei, W.W. Lin, Y.C. Yeh, S.M. Hwang, J.J. Wang, M.S. Tsai, Y. Chang,
cardiomyogenic differentiation of rat bone marrow mesenchymal stem cells, et al., Enhancement of cell retention and functional benefits in myocardial infarc-
Arch. Med. Res. 41 (2010) 497–505. tion using human amniotic-fluid stem-cell bodies enriched with endogenous
[67] H. Song, H.J. Hwang, W. Chang, B.W. Song, M.J. Cha, I.K. Kim, S. Lim, E.J. Choi, et al., ECM, Biomaterials 32 (2011) 5558–5567.
Cardiomyocytes from phorbol myristate acetate-activated mesenchymal stem cells [94] J.P. Karam, C. Muscari, C.N. Montero-Menei, Combining adult stem cells and poly-
restore electromechanical function in infarcted rat hearts, Proc. Natl. Acad. Sci. U. S. meric devices for tissue engineering in infarcted myocardium, Biomaterials 33
A. 108 (2011) 296–301. (2012) 5683–5695.
[68] J. Bartunek, J.D. Croissant, W. Wijns, S. Gofflot, A. de Lavareille, M. Vanderheyden, Y. [95] E. Ruvinov, T. Harel-Adar, S. Cohen, Bioengineering the infarcted heart by applying
Kaluzhny, N. Mazouz, et al., Pretreatment of adult bone marrow mesenchymal bio-inspired materials, J. Cardiovasc. Transl. Res. 4 (2011) 559–574.
stem cells with cardiomyogenic growth factors and repair of the chronically in- [96] L.L. Chiu, R.K. Iyer, L.A. Reis, S.S. Nunes, M. Radisic, Cardiac tissue engineering: cur-
farcted myocardium, Am. J. Physiol. Heart Circ. Physiol. 292 (2007) H1095–H1104. rent state and perspectives, Front. Biosci. 17 (2012) 1533–1550.
[69] D.A. Pijnappels, M.J. Schalij, A.A. Ramkisoensing, J. van Tuyn, A.A. de Vries, A. van [97] D. Simpson, H. Liu, T.H. Fan, R. Nerem, S.C. Dudley Jr., A tissue engineering ap-
der Laarse, D.L. Ypey, D.E. Atsma, Forced alignment of mesenchymal stem cells un- proach to progenitor cell delivery results in significant cell engraftment and im-
dergoing cardiomyogenic differentiation affects functional integration with cardio- proved myocardial remodeling, Stem Cells 25 (2007) 2350–2357.
myocyte cultures, Circ. Res. 103 (2008) 167–176. [98] J. Jin, S.I. Jeong, Y.M. Shin, K.S. Lim, Y.M. Lee, H.C. Koh, K.S. Kim, Transplantation of
[70] M. Gnecchi, Z. Zhang, A. Ni, V.J. Dzau, Paracrine mechanisms in adult stem cell sig- mesenchymal stem cells within a poly (lactide‐co‐ɛ‐caprolactone) scaffold im-
naling and therapy, Circ. Res. 103 (2008) 1204–1219. proves cardiac function in a rat myocardial infarction model, Eur. J. Heart Fail. 11
[71] R. Zamilpa, M.M. Navarro, I. Flores, S. Griffey, Stem cell mechanisms during left (2009) 147–153.
ventricular remodeling post-myocardial infarction: repair and regeneration, [99] K.L. Fujimoto, K. Tobita, W.D. Merryman, J. Guan, N. Momoi, D.B. Stolz, M.S. Sacks,
World J. Cardiol. 6 (2014) 610–620. B.B. Keller, et al., An elastic, biodegradable cardiac patch induces contractile
[72] S.H. Bhang, S.W. Cho, W.G. La, T.J. Lee, H.S. Yang, A.Y. Sun, S.H. Baek, J.W. Rhie, et al., smooth muscle and improves cardiac remodeling and function in subacute myo-
Angiogenesis in ischemic tissue produced by spheroid grafting of human adipose- cardial infarction, J. Am. Coll. Cardiol. 49 (2007) 2292–2300.
derived stromal cells, Biomaterials 32 (2011) 2734–2747. [100] H. Piao, J.S. Kwon, S. Piao, J.H. Sohn, Y.S. Lee, J.W. Bae, K.K. Hwang, D.W. Kim, et al.,
[73] H.J. Hwang, W. Chang, B.W. Song, H. Song, M.J. Cha, I.K. Kim, S. Lim, E.J. Choi, et al., Effects of cardiac patches engineered with bone marrow-derived mononuclear
Antiarrhythmic potential of mesenchymal stem cell is modulated by hypoxic envi- cells and PGCL scaffolds in a rat myocardial infarction model, Biomaterials 28
ronment, J. Am. Coll. Cardiol. 60 (2012) 1698–1706. (2007) 641–649.
[74] T. Kinnaird, E. Stabile, M.S. Burnett, C.W. Lee, S. Barr, S. Fuchs, S.E. Epstein, Marrow- [101] Y.D. Lin, M.L. Yeh, Y.J. Yang, D.C. Tsai, T.Y. Chu, Y.Y. Shih, M.Y. Chang, Y.W. Liu, et al.,
derived stromal cells express genes encoding a broad spectrum of arteriogenic cy- Intramyocardial peptide nanofiber injection improves postinfarction ventricular
tokines and promote in vitro and in vivo arteriogenesis through paracrine mecha- remodeling and efficacy of bone marrow cell therapy in pigs, Circulation 122
nisms, Circ. Res. 94 (2004) 678–685. (2010) S132–141.
[75] S.L. Zhao, Y.J. Zhang, M.H. Li, X.L. Zhang, S.L. Chen, Mesenchymal stem cells with [102] G. Dubois, V.F. Segers, V. Bellamy, L. Sabbah, S. Peyrard, P. Bruneval, A.A. Hagege,
overexpression of midkine enhance cell survival and attenuate cardiac dysfunction R.T. Lee, et al., Self-assembling peptide nanofibers and skeletal myoblast transplan-
in a rat model of myocardial infarction, Stem Cell Res. Ther. 5 (2014) 37. tation in infarcted myocardium, J. Biomed. Mater. Res. B Appl. Biomater. 87 (2008)
[76] A. Paul, A. Hasan, H.A. Kindi, A.K. Gaharwar, V.T. Rao, M. Nikkhah, S.R. Shin, D. 222–228.
Krafft, et al., Injectable graphene oxide/hydrogel-based angiogenic gene delivery [103] X.J. Cui, H. Xie, H.J. Wang, H.D. Guo, J.K. Zhang, C. Wang, Y.Z. Tan, Transplanta-
system for vasculogenesis and cardiac repair, ACS Nano 8 (2014) 8050–8062. tion of mesenchymal stem cells with self-assembling polypeptide scaffolds is
[77] G. Zhang, Y. Nakamura, X. Wang, Q. Hu, L.J. Suggs, J. Zhang, Controlled release of conducive to treating myocardial infarction in rats, Tohoku J. Exp. Med. 222
stromal cell-derived factor-1 alpha in situ increases c-kit + cell homing to the in- (2010) 281–289.
farcted heart, Tissue Eng. 13 (2007) 2063–2071. [104] H.D. Guo, G.H. Cui, H.J. Wang, Y.Z. Tan, Transplantation of marrow-derived cardiac
[78] Y. Zhang, W. Li, L. Ou, W. Wang, E. Delyagina, C. Lux, H. Sorg, K. Riehemann, et al., stem cells carried in designer self-assembling peptide nanofibers improves cardiac
Targeted delivery of human VEGF gene via complexes of magnetic nanoparticle- function after myocardial infarction, Biochem. Biophys. Res. Commun. 399 (2010)
adenoviral vectors enhanced cardiac regeneration, PLoS One 7 (2012) e39490. 42–48.
[79] E.J. Battegay, J. Rupp, L. Iruela-Arispe, E.H. Sage, M. Pech, PDGF-BB modulates endo- [105] H.S. Chen, C. Kim, M. Mercola, Electrophysiological challenges of cell-based myo-
thelial proliferation and angiogenesis in vitro via PDGF beta-receptors, J. Cell Biol. cardial repair, Circulation 120 (2009) 2496–2508.
125 (1994) 917–928. [106] B.J. Kang, H. Kim, S.K. Lee, J. Kim, Y. Shen, S. Jung, K.S. Kang, S.G. Im, et al., Umbilical-
[80] D.F. Bowen-Pope, T.W. Malpass, D.M. Foster, R. Ross, Platelet-derived growth factor cord-blood-derived mesenchymal stem cells seeded onto fibronectin-immobilized
in vivo: levels, activity, and rate of clearance, Blood 64 (1984) 458–469. polycaprolactone nanofiber improve cardiac function, Acta Biomater. 10 (2014)
[81] A. Luttun, M. Tjwa, L. Moons, Y. Wu, A. Angelillo-Scherrer, F. Liao, J.A. Nagy, A. 3007–3017.
Hooper, et al., Revascularization of ischemic tissues by PlGF treatment, and inhibi- [107] K.H. Shin, Y. Jang, B.S. Kim, J. Jang, S.H. Kim, Highly conductive reduced graphene
tion of tumor angiogenesis, arthritis and atherosclerosis by anti-Flt1, Nat. Med. 8 oxide produced via pressure-assisted reduction at mild temperature for flexible
(2002) 831–840. and transparent electrodes, Chem. Commun. 49 (2013) 4887–4889.
[82] S. Lehrman, Virus treatment questioned after gene therapy death, Nature 401 [108] P.R. Bandaru, Electrical properties and applications of carbon nanotube structures,
(1999) 517–518. J. Nanosci. Nanotechnol. 7 (2007) 1239–1267.
[83] H. Yin, R.L. Kanasty, A.A. Eltoukhy, A.J. Vegas, J.R. Dorkin, D.G. Anderson, Non-viral [109] M. Shevach, S. Fleischer, A. Shapira, T. Dvir, Gold nanoparticle-decellularized ma-
vectors for gene-based therapy, Nat. Rev. Genet. 15 (2014) 541–555. trix hybrids for cardiac tissue engineering, Nano Lett. 14 (2014) 5792–5796.
[84] M. Neu, D. Fischer, T. Kissel, Recent advances in rational gene transfer vector design [110] N.J. Severs, A.F. Bruce, E. Dupont, S. Rothery, Remodelling of gap junctions and
based on poly(ethylene imine) and its derivatives, J. Gene Med. 7 (2005) connexin expression in diseased myocardium, Cardiovasc. Res. 80 (2008)
992–1009. 9–19.
[85] Y.C. Huang, T.J. Liu, Mobilization of mesenchymal stem cells by stromal cell- [111] R. Ravichandran, R. Sridhar, J.R. Venugopal, S. Sundarrajan, S. Mukherjee, S.
derived factor-1 released from chitosan/tripolyphosphate/fucoidan nanoparticles, Ramakrishna, Gold nanoparticle loaded hybrid nanofibers for cardiogenic differen-
Acta Biomater. 8 (2012) 1048–1056. tiation of stem cells for infarcted myocardium regeneration, Macromol. Biosci. 14
[86] J.D. Abbott, Y. Huang, D. Liu, R. Hickey, D.S. Krause, F.J. Giordano, Stromal cell- (2014) 515–525.
derived factor-1alpha plays a critical role in stem cell recruitment to the heart [112] J. Park, Y.S. Kim, S. Ryu, W.S. Kang, S. Park, J. Han, H.C. Jeong, B.H. Hong, et al.,
after myocardial infarction but is not sufficient to induce homing in the absence Graphene potentiates the myocardial repair efficacy of mesenchymal stem cells
of injury, Circulation 110 (2004) 3300–3305. by stimulating the expression of angiogenic growth factors and gap junction pro-
[87] A. Schober, S. Knarren, M. Lietz, E.A. Lin, C. Weber, Crucial role of stromal cell- tein, Adv. Funct. Mater. 25 (2015) 2590–2600.
derived factor-1 alpha in neointima formation after vascular injury in [113] O.C. Compton, S.T. Nguyen, Graphene oxide, highly reduced graphene oxide, and
apolipoprotein-E-deficient mice, J. Vasc. Res. 41 (2004) 104-104. graphene: versatile building blocks for carbon-based materials, Small 6 (2010)
[88] V.F. Segers, T. Tokunou, L.J. Higgins, C. MacGillivray, J. Gannon, R.T. Lee, Local deliv- 711–723.
ery of protease-resistant stromal cell derived factor-1 for stem cell recruitment [114] M.J. Webber, X. Han, S.N. Murthy, K. Rajangam, S.I. Stupp, J.W. Lomasney, Capturing
after myocardial infarction, Circulation 116 (2007) 1683–1692. the stem cell paracrine effect using heparin-presenting nanofibres to treat cardio-
[89] M.E. Padin-Iruegas, Y. Misao, M.E. Davis, V.F. Segers, G. Esposito, T. Tokunou, vascular diseases, J. Tissue Eng. Regen. Med. 4 (2010) 600–610.
K. Urbanek, T. Hosoda, et al., Cardiac progenitor cells and biotinylated [115] J. Park, S. Park, S. Ryu, S.H. Bhang, J. Kim, J.K. Yoon, Y.H. Park, S.P. Cho, et al.,
insulin-like growth factor-1 nanofibers improve endogenous and exoge- Graphene-regulated cardiomyogenic differentiation process of mesenchymal
nous myocardial regeneration after infarction, Circulation 120 (2009) stem cells by enhancing the expression of extracellular matrix proteins and cell
876–887. signaling molecules, Adv. Healthcare Mater. 3 (2014) 176–181.
28 J. Han et al. / Advanced Drug Delivery Reviews 95 (2015) 15–28

[116] Y. Miyahara, N. Nagaya, M. Kataoka, B. Yanagawa, K. Tanaka, H. Hao, K. Ishino, H. [120] D. Song, X. Liu, R. Liu, L. Yang, J. Zuo, W. Liu, Connexin 43 hemichannel regulates
Ishida, et al., Monolayered mesenchymal stem cells repair scarred myocardium H9c2 cell proliferation by modulating intracellular ATP and [Ca2+], Acta Biochim.
after myocardial infarction, Nat. Med. 12 (2006) 459–465. Biophys. Sin. 42 (2010) 472–482.
[117] H. Okura, A. Matsuyama, C.M. Lee, A. Saga, A. Kakuta-Yamamoto, A. Nagao, N. [121] T.J. Lee, S. Park, S.H. Bhang, J.K. Yoon, I. Jo, G.J. Jeong, B.H. Hong, B.S. Kim, Graphene
Sougawa, N. Sekiya, et al., Cardiomyoblast-like cells differentiated from human ad- enhances the cardiomyogenic differentiation of human embryonic stem cells,
ipose tissue-derived mesenchymal stem cells improve left ventricular dysfunction Biochem. Biophys. Res. Commun. 452 (2014) 174–180.
and survival in a rat myocardial infarction model, Tissue Eng. 16 (2010) 417–425. [122] X.T. Shi, H.X. Chang, S. Chen, C. Lai, A. Khademhosseini, H.K. Wu, Regulating cellular
[118] Y. Akiyama, A. Kikuchi, M. Yamato, T. Okano, Ultrathin poly(N-isopropylacrylamide) behavior on few-layer reduced graphene oxide films with well-controlled reduc-
grafted layer on polystyrene surfaces for cell adhesion/detachment control, Langmuir tion states, Adv. Funct. Mater. 22 (2012) 751–759.
20 (2004) 5506–5511.
[119] A. Kikuchi, T. Okano, Nanostructured designs of biomedical materials: applications
of cell sheet engineering to functional regenerative tissues and organs, J. Control.
Release 101 (2005) 69–84.

You might also like