You are on page 1of 10

JOURNAL OF TISSUE ENGINEERING AND REGENERATIVE MEDICINE RESEARCH ARTICLE

J Tissue Eng Regen Med (2015)


Published online in Wiley Online Library (wileyonlinelibrary.com) DOI: 10.1002/term.1995

Three-dimensional functional human myocardial


tissues fabricated from induced pluripotent
stem cells
Hyoe Komae1,2, Hidekazu Sekine2, Izumi Dobashi2, Katsuhisa Matsuura2, Minoru Ono1,
Teruo Okano2 and Tatsuya Shimizu2*
1
Department of Cardiac Surgery, University of Tokyo, Japan
2
Institute of Advanced Biomedical Engineering and Science, Tokyo Womens Medical University, Japan

Abstract
The most radical treatment currently available for severe heart failure is heart transplantation;
however, the number of donor hearts is limited. A better approach is to make human cardiac tissues.
We developed an original cell sheet-based tissue-engineering technology to fabricate human cardiac
tissue by layering myocardial cell sheets. Human induced pluripotent stem (iPS) cells were differen-
tiated into cardiomyocytes to fabricate cardiomyocyte sheets. Initially, three-layer human iPS cardio-
myocyte (hiPSCM) sheets were transplanted on subcutaneous tissues of nude rats. Next, to fabricate
thicker tissue, three-layer sheets were transplanted on one day, then additional three-layer sheets
were transplanted onto them the following day, after the rst sheets were vascularized. On day 3,
the nal three-layer sheets were again transplanted, creating a nine-layer graft (multi-step transplan-
tation procedure). In the last step, six-layer sheets were transplanted on fat tissues of the inguinal
portion, which were subsequently resected together with the femoral arteries and veins to make
transplantable grafts with connectable vessels. They were then transplanted ectopically to the neck
portion of other rats by anastomosing vessels with the hosts jugular arteries and veins. Transplanted
three-layer hiPSCMs were beating and, histologically, showed a cardiac muscle-like structure with
vascular systems. Moreover, transplanted hiPSCMs proliferated and matured in vivo. Signicantly
thicker tissues were fabricated by a multi-step transplantation procedure. The ectopically
transplanted graft survived and continued to beat. We succeeded in fabricating functional human
cardiac tissue with cell sheet technology. Transplanting this cardiac tissue may become a new
treatment option for severe heart failure. Copyright 2015 John Wiley & Sons, Ltd.

Received 31 July 2014; Revised 4 November 2014; Accepted 9 December 2014

Keywords cell sheet; iPS cells; cardiomyocyte; cardiac tissue; transplantaion; heart failure

1. Introduction In cardiac regenerative medicine, cell injection


methods were the rst to be commonly applied (Chugh
et al., 2012; Hare et al., 2012; Makkar et al., 2012).
Recent progress in the eld of regenerative medicine
Some of these injection methods did show improve-
has been remarkable, and many new therapeutic
ments in the ejection fraction and reduction of scar size
methods have been developed. Within this eld, cell-
but problems remain, such as low cell survival rates
based regenerative therapy is thought to be the most
and the difculty of keeping the cells in the designated
promising.
areas.
These problems have been overcome by creating
three-dimensional (3D) tissues from single-source cells,
*Correspondence to: Tatsuya Shimizu, Institute of Advanced Bio-
medical Engineering and Science, TWIns, Tokyo Womens Medi- and this method will become one of the most effective
cal University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo 162-8666, treatments. Since (Langer and Vacanti, 1993) succeeded
Japan. E-mail: shimizu.tatsuya@twmu.ac.jp in fabricating ear cartilage using a biodegradable

Copyright 2015 John Wiley & Sons, Ltd.


H. Komae et al.

polymer scaffold, and suggested the concept of tissue Use of Laboratory Animals prepared by the Institute of
engineering, scaffold-based tissue engineering has be- Laboratory Animal Resources (ILAR).
come predominant and has already been used in clinical
applications (Crowley, 2013; Nemeno-Guanzon et al.,
2012; Tuan et al., 2013; Yildirimer et al., 2012). How- 2.1. Cardiac differentiation from human iPS cells
ever, cell-dense tissues, such as heart, were difcult to and fabrication of human myocardial tissue
reconstruct because the seeded cells were isolated, grafts
creating a lower cell density, which became even lower
after the scaffold biodegraded. HiPS cells were purchased from RIKEN (253G1, Tsukuba,
On the other hand, we have originally developed a Japan) and cultured using a bioreactor system to induce
cell sheet-based tissue-engineering technology that re- differentiation into hiPSCMs, as previously reported
quires no scaffolds. By culturing cells on temperature- (Matsuura et al., 2012), with some modications
responsive culture dishes and simply lowering the tem- (Figure 1A). To make cell sheets, 2 106 differentiated
perature, the conuently cultured cells are released in hiPSCMs in this method were seeded on 35 mm
a sheet form (Okano et al., 1993). By stacking the cell temperature-responsive dishes (UpCell, CellSeed, Tokyo,
sheets, we can successfully make scaffold-free, 3D Japan) in Dulbeccos modied Eagles medium (DMEM;
tissues with cellcell adhesive molecules (Shimizu Sigma-Aldrich, St. Louis, MO, USA) supplemented with
et al., 2002). 10% fetal bovine serum (FBS) and cultured until they
We have reported success in forming thick, beating grew to conuency. The culture dishes were placed in an
heart tissue with vascular networks by layering neonatal incubator set at 20C; the hiPSCM sheets detached spon-
rat cardiomyocyte sheets in vivo (Shimizu et al., 2006a; taneously within 1 h. A detached sheet was spread at,
Shimizu et al., 2006b). We believe that human car- the medium was aspirated and the dish was held at 37C
diomyocytes can also be used to create cell sheets, for 1 h to allow the cell sheet to adhere. Another detached
which can reconstruct thick human myocardial tissues hiPSCM sheet was then transferred and stacked onto the
when they were layered, opening new treatment options rst sheet, using a pipette, to make two layers. The re-
in cardiac regenerative medicine. Recently, techniques maining medium was aspirated and the dish was held at
for differentiating human stem cells, such as embryonic 37C for another 1 h. An identical procedure was repeated
stem (ES) cells or induced pluripotent stem (iPS) cells, to layer a third sheet, thus fabricating the three-layer
into human cardiomyocytes have now been established. hiPSCM sheets (Figure 1B).
Applying these techniques, a human heart graft can
now be fabricated.
Many groups, including ourselves, have tried to use this 2.2. Transplantation of engineered human
new tissue-engineering technology in clinical practice. An- myocardial tissue grafts
other group has reported that cardiac tissue sheets made
up of human cardiomyocytes differentiated from iPS cells, Male Fischer 344 athymic nude rats (Charles River Japan,
endothelial cells and vascular mural cells were transplanted Tokyo, Japan), aged 45 weeks, were anaesthetized by
onto rat infarcted hearts, which signicantly improved car- 2% isourane inhalation. An L-shaped incision was made
diac function (Masumoto et al., 2014). Moreover, we are in the dorsal skin, exposing the dorsal subcutaneous tissue.
now trying to fabricate a thick human heart tissue graft to Three-layer hiPSCM sheets on temperature-responsive
strengthen this effect, or support cardiac contraction, which dishes were detached by lowering the temperature to 20
will be an innovative treatment for heart failure and is our C, and washed with lactated Ringers solution. The hiPSCM
ultimate goal. sheets were lifted onto a sterile polypropylene support
In this research, cardiomyocytes differentiated from sheet (2 1.5 cm), transplanted onto the subcutaneous tis-
human induced pluripotent stem (hiPS) cells were sue and covered with a 0.5 mm-thick silicone membrane
made into cell sheets. Stacking these hiPS cardiomyo- (Unique Medical, Tokyo, Japan) to prevent adhesion to
cyte (hiPSCM) sheets in vivo, we tried to make thick the skin, then the incisions were closed (Shimizu et al.,
functional human cardiac tissues and to transplant this 2006b). The transplanted portions were observed 2 weeks
tissue ectopically, in order to improve the methods for later (n = 5) or every month up to 6 months (n = 8)
fabricating thick functional human cardiac tissues and (Figure 1C). When grafts of hiPSCM sheets with more than
their ectopic transplantation. three layers were needed, an initial three-layer hiPSCM
sheet was transplanted and, after 1 day, a second three-
layer sheet was placed on the rst transplanted sheet, then
a third three-layer hiPSCM sheet on the following day.
Allowing 1 day between each transplantation encouraged
2. Methods vascularization, which supports the survival of the nal
hiPSCM tissue (Figure 1D). In this multi-step transplanta-
All animal experiments were performed according to the tion procedure, hiPSCM sheets were repeatedly stacked
Guidelines of Tokyo Womens Medical University on Ani- three times to make nine layers and were observed 2 weeks
mal Use, and consistent with the Guide for the Care and later (n = 4) (Figure 1C).

Copyright 2015 John Wiley & Sons, Ltd. J Tissue Eng Regen Med (2015)
DOI: 10.1002/term
3D functional human myocardial tissues from iPS cells

Figure 1. Methods of hiPSCM sheet transplantation. hiPS cells (blue) were cultured using a bioreactor system to induce differentia-
tion into hiPSCMs in procedures, as shown. (A) About 8090% of differentiated cells were cardiomyocytes (red) and the rest were sup-
posed to be mural cells (yellow). (B) Differentiated cells were seeded on temperature-responsive dishes and cultured until they grew
to conuency; then, by lowering the temperature, the hiPSCM sheets spontaneously detached and were stacked to three layers
in vitro, then (C) the sheets were transplanted in vivo. (D) By stacking over three layers, with repeated transplantation of layered cell
sheets at 1 day intervals (which we call a multi-step transplantation procedure), we were able to overcome the thickness limitation

2.3. Fabrication of grafts with connectable Frozen cross-sections of the resected grafts were sec-
vessels and their ectopic transplantation tioned sagittally into 610 m slices and haematoxylin
and eosin (H&E) staining was performed by conventional
Male F344 nude rats, aged > 8 weeks, weight 300350 g, methods. For immunohistochemistry, mouse anti-troponin
were anaesthetized and incisions were made in the ingui- T antibodies (Thermo Fisher Scientic, Waltham, MA,
nal portion. A six-layer graft was made, using the multi- USA), rabbit polyclonal anti-CD31 antibodies (Thermo
step procedure, on the fat tissue perfused with a branch Fisher Scientic), mouse anti--actinin antibodies
of the femoral artery and vein (n = 2). Two weeks later, (Sigma-Aldrich), rabbit monoclonal anti-Ki67 antibodies
the fat tissue with the six-layer construct was resected, (Abcam) and rabbit polyclonal anti-Histone H3 antibodies
with the femoral artery and vein, after injecting heparin (Abcam) were used for primary antibodies. Fluorescently
(1000 U). Subsequently, the graft was transplanted into labelled secondary antibodies (AlexaFluor 568-labelled
the neck of another F344 rat (300350 g) by anastomos- goat anti-mouse IgG antibodies, AlexaFluor 488-labelled
ing vessels with the hosts jugular artery and vein, using goat anti-mouse IgG antibodies, AlexaFluor 488-labelled
a cuff technique. The transplanted graft was observed 1 goat anti-rabbit IgG antibodies (Molecular Probes, Eugene,
week later (Figure 1C). OR, USA) were used for secondary antibodies.
The sections were nally counterstained with anti-fade
solution (ProLong Gold Antifade Reagent with DAPI, Molecu-
2.4. Histological analysis lar Probes) and were observed using confocal laser scanning
microscopy (FV1200 IX83, Olympus, Tokyo, Japan).
Rats were anaesthetized as described and transplanted
sites together with underlying tissues were resected. In
some cases, 1 mg lectin (DyLight 594 Labelled Tomato 2.5. Electron microscopy analysis
Lectin, DyLight 488 Labelled Tomato Lectin, Abcam, Cam-
bridge, UK) was injected into the penile venous plexus. Transplanted grafts were harvested and specimens were
The rats were then sacriced under adequate anaesthesia. xed and processed by Tokai Electron Microscopy Inc.

Copyright 2015 John Wiley & Sons, Ltd. J Tissue Eng Regen Med (2015)
DOI: 10.1002/term
H. Komae et al.

(Nagoya, Japan) for electron microscopy analysis. The 3. Results


specimens were embedded into 100% resin and polymer-
ized to make blocks. The blocks were then ultra-thinly 3.1. Thick human cardiac tissue fabrication by
sectioned at 70 nm and stained with 2% uranyl acetate.
the multi-step transplantation procedure
They were observed using a transmission electron micro-
scope (JEM-1200EX; Jeol Ltd, Tokyo, Japan) at an acceler-
The transplanted three-layer hiPSCM sheets were still beat-
ation voltage of 80 kV. Digital images were taken with a
ing macroscopically after 2 weeks. Synchronized electrical
CCD camera (Veleta; Olympus Soft Imaging Solutions
potentials were detected (Figure 2A). The results of uores-
GmbH, Mnster, Germany).
cent immunostaining suggested that the transplanted
hiPSCM sheets had survived as cardiac tissues, with sarco-
2.6. Electrophysiological analysis mere structures and microvascular networks (Figure 2E, G)
connecting with the hosts circulatory system (Figure 2I).
Transplantation sites were opened, and two microelec- The mapping indicated that there was no arrhythmia
trodes (29 G in diameter, ADInstruments, New South (see supporting information, Video S1). In three cases of
Wales, Australia) were positioned over the grafts. Electrical nine-layer hiPSCM transplantation, macroscopic beating
potentials were amplied by bioelectric ampliers (UA102, and synchronized electrical potentials were detected 2 weeks
Unique Medical) and recorded using a data acquisition sys- later (Figure 2B). The uorescent immunostaining also
tem (ML870, PowerLab 8/30, ADInstruments). At the same showed that the cardiac tissues with sarcomere structures
time, the hosts electrocardiogram and body pressure were and microvascular networks had survived (Figure 2F, H).
recorded. Some vessels were observed penetrating the transplanted
graft (Figure 2J).
Measuring the thickness of the transplanted grafts,
2.7. Membrane potential mappings the nine-layer transplanted sheets were signicantly
thicker (359 161 m, n = 4) than the three-layer
Grafts were made as ectopic transplantation models. Three sheets (165 40 m, n = 5); p < 0.01 (Figure 2C, D).
weeks later, the resected graft was placed in a tissue cham-
ber (ABLE). First, the graft was perfused with 2.5 ml culture
medium containing 2 mM voltage-sensitive dye RH237 3.2. Long-term observation of three-layer
(Molecular Probes) dissolved in dimethyl sulphoxide and transplantation
5 M blebbistatin (Tocris, Ellisville, MO, USA) to inhibit con-
traction, at 50 l/min. Second, it was perfused with me- Macroscopic beating and synchronized electrical poten-
dium at the same ow rate in the bioreactor system, tials were conrmed at 6 months after transplantation
where O2 levels were maintained at 20%, CO2 levels at (Figure 3A). The beating appeared strong (see supporting
5%, pH level at 7.4 and temperature at 37C. information, Video S2) and the grafts were well-
An hour after RH 237 perfusion, the graft was excited with vascularized (Figure 3B). The results of uorescent immu-
an LED light source (LEX2-G, BrainVision, Tokyo, Japan) at nostaining indicated that the structure of the cardiac tis-
532 nm, and uorescent images were imaged with a dichroic sues contained sarcomere structures and a microvascular
mirror (650 nm cut-off wavelength). Emission uorescence network (Figure 3C); in addition, gap junctions between
of 715 nm was collected using a CMOS camera (MiCAM cardiac cells were observed at 6 months (Figure 3D).
Ultima, BrainVision) and a Tandem-lens scope (BrainVision) The fabricated tissue was more cell-dense at 6 months
at 1 ms/frame and 0.16 mm2/pixel. Magnication of the ob- than the grafts at 2 weeks after transplantation, which im-
jective lens was 0.63 power (Choi and Salama, 2000). The plied that hiPSCMs were proliferating after transplanta-
data were analyzed using BV_Ana software (Brainvision) to tion. To see the transplanted hiPSCMs proliferation,
process it, using a spatial lter for activation time analysis uorescent immunostaining for Ki67 and phosphor-
and activation mapping. histone H3 (PPH3) was performed. From the results,
many hiPSCMs were proliferating at 2 weeks after trans-
plantation (Figure 3G, H); however, hiPSCMs had
2.8. Video capture
stopped proliferation by 6 months (Figure 3E, F).
Macroscopic images were recorded using a digital video
camera (DCR-TRV900, Sony, Tokyo, Japan) and edited
with Adobe Premiere Pro software. 3.3. Maturation of transplanted hiPSCMs

From the electron microscopy images, the hiPSCM


2.9. Data analysis sheets had poor myobrils (Figure 4A, B), but myobrils
had fused and elongated as time passed and desmo-
Data are expressed as mean SD. Statistical analysis was somes could be seen at 6 months (Figure 4E). Moreover,
performed using Students unpaired t-test; p < 0.05 was the number of mitochondria in the cytoplasm increased
considered signicant. in the transplanted grafts. Either a sarcoplasmic

Copyright 2015 John Wiley & Sons, Ltd. J Tissue Eng Regen Med (2015)
DOI: 10.1002/term
3D functional human myocardial tissues from iPS cells

Figure 2. Human cardiac tissues fabricated from hiPSCM sheets transplanted in vivo; results of three-layer hiPSCM sheet transplantation
after 2 weeks (A, C, E, G) and of nine-layer sheets (B, D, F, H). Electrical potential changes (A, B; blue) are regular and completely independent
from host ECG (red) or breathing (green). The thickness of the three-layer graft was 165 40 m (C; n = 5; *) and 359 161 m in the nine-
layer cases (D; n = 4; **). Fluorescent immunostaining shows cardiac tissues with microvascular systems (E, F; red, cTnT; green, CD31) and
the striated structure of sarcomeres (G, H; red, -actinin). Grafts were perfused from the host circulatory systems. Fabricated graft was
stained by red lectin injected into the host (I, arrows). Some vessels are penetrating the transplanted hiPSCM sheets (J; arrows)

Copyright 2015 John Wiley & Sons, Ltd. J Tissue Eng Regen Med (2015)
DOI: 10.1002/term
H. Komae et al.

Figure 3. Results at 6 months after transplantation and hiPSCM proliferation. Electrical potential change (A; blue) was regular and
completely independent from host ECG (red) and breathing (green). Transplanted hiPSCM sheets (B; circled with arrows) appear
to be vascular-rich tissue. HiPSCM sheets became cell-dense cardiac tissue with vascular networks (C; red, cTnT; green, CD31) and
gap junctions (D; red, -actinin; green, connexin43). While hiPSCMs were proliferating (G; red, cTnT; green, Ki67: H; red, cTnT; green,
PHH3) at 2 weeks after transplantation, proliferation stopped at 6 months (E; red, cTnT; green, Ki67: F; red, cTnT; green, PHH3)

reticulum or a T-tube was observed around a myobril transplanted in one step, central necrosis will develop,
at 6 months (Figure 4F). and the thickness of the construct could not exceed 80
m. Looking at the lower portions of the constructs that
did survive, a well-organized vascular network was
3.4. Successful ectopic transplantation of formed, which indicates perfusion from the host
vascularized tissue (Shimizu et al., 2006b). Therefore, we hypothesized that
multiple three-layer sheets could be transplanted if it
The grafts resected from the inguinal portion were beating, was onto a vascularized construct, which was veried.
and the synchronized electric potentials were measured It became clear that three-layer sheets can be layered
(Figure 5A). One week after ectopic transplantation, the at 1 day intervals; moreover, the resulting six-layer con-
grafts were also beating (Figure 5B). Histological study struct also developed a functional vascular network,
showed human cardiac tissue with microvascular networks which enabled more sheets to be transplanted. In this
that were perfused from the hosts circulatory system way, repeated transplantation of layered cell sheets af-
(Figure 5C, D). ter an interval of 1 day produced neovascularization
within the previously implanted graft and overcame
the limit of thickness (Figure 1D, multi-step transplanta-
4. Discussion tion procedure).
If thick human cardiac tissues are to be fabricated, a
This is the rst report of thick functional human myocar- large number of hiPSCM sheets must be layered. There-
dial tissue to be successfully fabricated from hiPSCM fore, we needed to clarify whether our multi-step trans-
sheets in vivo. We succeeded in reconstructing functional plantation procedure could be used with hiPSCM sheets.
human myocardial tissue thicker than 350 m from Initially, we conrmed that transplanted three-layer
hiPSCM sheets by using a multi-step transplantation pro- iPSCM sheets did survive, and that functional vascular
cedure, and transplanted this fabricated cardiac tissue networks connected to the hosts circulatory system
ectopically. were formed in the sheets. Moreover, the electric mem-
brane potential of the fabricated grafts proved to ow in
one direction, like native human cardiac tissues. Then,
4.1. Fabrication of thick human cardiac tissue additional hiPSCM sheets could be transplanted onto
from hiPSCM sheets the rst vascularized sheets, and they survived. By re-
peating this, hiPSCM sheets were stacked up to nine
To fabricate thick tissue, vascularization within the con- layers. This was largely because the myocardial tissues
struct is critical. From our previous study we learned had formed functional microvascular networks within
that when more than three layers of cell sheets are the transplanted layers. Therefore, we can now say that

Copyright 2015 John Wiley & Sons, Ltd. J Tissue Eng Regen Med (2015)
DOI: 10.1002/term
3D functional human myocardial tissues from iPS cells

Figure 4. Results of electron microscopy. Low-powered (A, C, E) and high-powered (B, D, F) images of hiPSCM sheets (A, B) at 2
weeks (C, D) and 6 months (E, F). The cells are myobril-poor and sparse in the hiPSCM sheets (B), but the myobrils became dense,
wide and elongated over time, and desmosomes were formed at 6 months (E; blank arrows). Mitochondria increased in number (A, C,
E; arrows); arrowhead indicates sarcoplasmic reticulum or T-tubes around myobrils (F)

much thicker human myocardial tissue can be fabricated cardiomyocyte sheets would contain broblasts or other
in vivo by layering hiPSCM sheets using a multi-step factors, which would let cells bind as they were. HiPSCMs
procedure. are rather young cells, approximately corresponding to
human embryonic cardiomyocytes, which freely prolifer-
ate. It is possible that cell adhesion or assembly will be
4.2. HiPSCMs after transplantation in vivo weaker because hiPSCM sheets contain a higher propor-
tion of cardiomyocytes (8090%) than native tissue. We
Two weeks after transplantation, the fabricated tissue also observed transplanted sheets up to 6 months and
from three-layer hiPSCM sheets had increased in thick- found that the beating became more obvious and the fab-
ness to 165 m, which seemed inconsistent with our ricated cardiac tissues became more cell-dense over time.
previous report that layered sheets > 100 m could At 6 months, cell proliferation had almost stopped, like
not be transplanted at one time. However, our hypothe- native human cardiomyocytes after birth.
sis that hiPSCMs were proliferating after transplantation Previous infant autopsy reports say that human
in vivo now makes sense. HiPSCM sheets survived by cardiomyocytes proliferate during gestation and that pro-
developing functional microvascular networks, and liferation stops at approximately 23 weeks post partum
hiPSCMs could keep proliferating (Figure 3G, H). There- (Austin et al., 1995; Huttenbach et al., 2001; Mayhew
fore, transplanted three-layer sheets exceeded the previ- et al., 1997; Mayhew et al., 1998). Moreover, the rate of
ous thickness limit. proliferating nuclei is highest during weeks 1228 of ges-
However, the tissue was cell-sparse and the orientation tation, and after 28 weeks it decreases (Huttenbach
was not orderly compared to that of the infant rat cardio- et al., 2001). The human infant heart is formed at about
myocyte sheet (Shimizu et al., 2002) (Figure 2C, D). This 5 weeks of gestation and, if the hiPSCMs correspond to na-
is probably because the infant rat cardiomyocytes were al- tive cardiomyocytes, they should proliferate for 2 weeks
ready mature to some degree, and infant rat after transplantation. Six months after transplantation

Copyright 2015 John Wiley & Sons, Ltd. J Tissue Eng Regen Med (2015)
DOI: 10.1002/term
H. Komae et al.

Figure 5. Ectopic transplantation: 2 weeks later, the transplanted six-layer hiPSCM sheets are beating (A; blue, graft potential) and
after 1 week the ectopically transplanted graft was also beating (B; blue). Histologically, the graft is perfused from the host circula-
tory system (C; arrow, penetrating vessel: D; arrows; red, CD31; green, lectin)

would then make them equivalent to around 30 weeks the cardiomyocytes had matured in structure and
gestation, when the native cardiomyocytes are proliferat- became similar to those of an adult (Sako, 1971a, 1971b,
ing less. 1971c, 1971d).
The electron microscopy results suggested that In this study, we succeeded in fabricating rather mature
hiPSCMs matured in vivo as cardiomyocytes; myobrils cardiac tissues with mature myobrils and elongated sar-
became dense, wide and elongated and the mitochondria comeres in vivo, although they were not as mature com-
increased in number over time (Figure 4A, C, E). More- pared with human heart tissues. One of our goals is to
over, cardiac-specic intracellular structures, such as the fabricate mature myocardial tissue with strong contractile
sarcoplasmic reticulum or T-tubes, could be seen at 6 forces. To achieve this goal, newer methods or factors will
months (Figure 4F), whereas they could not be detected be required.
at 2 weeks. HiPSCMs cultured in vitro also become more
mature to some degree, but they matured even more if
cultured in vivo (see supporting information, Figure S1). 4.3. Ectopic transplantation
Myobrils were being fused and elongated in the case of
either in vitro culture or in vivo, but the density of sarco- If thick human cardiac tissue were fabricated on the
mere units in hiPSCMs cultured in vivo were obviously intended portion, for example on the heart surface, it
higher. Intracellular structures of hiPSCMs cultured would require very invasive polysurgery, which is imprac-
in vivo were also more developed. Therefore, we can say tical. Therefore, the tissue should be fabricated in an-
that hiPSCMs were well suited for growing in vivo. other, less invasive, location and then transplanted
In the rat experiments, reasonably mature structures ectopically.
could be seen only 3 weeks after infant rat cardiomyocyte In the eld of clinical plastic surgery, various tissues are
sheets were transplanted (Shimizu et al., 2002), which reconstructed using free ap grafts by anastomosing
might support the previous hypothesis that infant rat nearby vessels with microsurgery to substitute defective
cardiomyocytes were initially more mature than the dif- tissue due to congenital malformation, surgical resection
ferentiated hiPSCMs. or traumatic injury (Brown and Shaw, 2010; Cannon
From the previous report, intracellular structures such et al., 2012; Mohan et al., 2013; Wong and Wei, 2010).
as myobrils, desmosomes and secretary granules were We thought that if our fabricated tissue could be
formed by fetal month 2, and mitochondria increased transplanted like a free ap, it would survive and main-
in number from at about fetal month 3. At fetal month 7, tain its function. Therefore, successful results of ectopic

Copyright 2015 John Wiley & Sons, Ltd. J Tissue Eng Regen Med (2015)
DOI: 10.1002/term
3D functional human myocardial tissues from iPS cells

transplantation in this study demonstrated that our hy- We successfully fabricated human cardiac tissue by
pothesis might be practical. When intending to transplant transplanting hiPSCM sheets in vivo. If this method con-
fabricated tissues onto the heart surface, graft arteries tinues to develop and fabricated tissues can be scaled up,
used in coronary artery bypass surgery can be anasto- we may be able to fabricate human cardiac tissue or
mosed with the graft. If the main artery is long enough, even a human heart itself in the future. Although further
it can be anastomosed from the aorta, like a great saphe- studies will be required, transplantation of this new fab-
nous vein graft. The main vein can also be anastomosed ricated cardiac tissue may become a new treatment op-
with the accompanying vein, or directly with the right tion for severe heart failure that will rescue more
atrium, so as not to create a shunt. patients.

4.4. Future clinical use


Conict of interest
Although there are problems such as the need for a large
number of cells, and the impractical process of layering T.O. is a founder and director of the board of CellSheed
that requires polysurgery in vivo, many new methods are Inc., licensing technologies and patents from Tokyo
under development that may solve these challenges. For Womens Medical University; and T.O. and T.S. are share-
example, many researchers are developing new, large- holders of CellSeed Inc.
scale, cell-culture systems (Chen et al., 2014; Matsuura
et al., 2012) and several procedures are being substituted
with ex vivo ones using bioreactor systems (Sakaguchi Acknowledgements
et al., 2013; Sekine et al., 2013). When we overcome these
challenges, this method will provide groundbreaking We thank Allan Nisbet for his useful comments and editing as-
treatment procedures. sistance. This study was supported by grants from the Japan
This new technique may resolve many of the problems Society for the Promotion of Science (JSPS) through the
Funding Programme for World-leading Innovative R&D on Sci-
with treatments currently available for severe heart fail-
ence and Technology (FIRST Programme), initiated by the
ure. When myocardial tissues or even hearts fabricated
Council for Science and Technology Policy (CSTP). We report
from iPS cells become available, they will be made from grants from: CSTEC, sponsored by MEXT during the conduct
human cells and perhaps even the patients own cells. of the study; Hitachi Ltd, Dai Nippon Printing Co. Ltd,
We plan to combine current therapeutic methods with CellSheed Inc., Olympus Corp., Terumo Corp., Nihon Kohden
fabricated human cardiac tissue, which will be able to re- Corp., Asahi Kasei Corp., Panasonic Corp., the National Insti-
duce many of the adverse effects to provide more effective tute of Material Science, Nikon Corp. and Kowa Co. Ltd out-
and safer treatments. side the submitted work.

References
Austin A, Fagan DG, Mayhew TM. 1995; A Crowley C, Wong JM, Fisher DM, et al. 2013; A vascular cells for cardiac regeneration.
stereological method for estimating the to- systematic review on preclinical and clinical Sci Rep 4: 6716.
tal number of ventricular myocyte nuclei studies on the use of scaffolds for bone repair Matsuura K, Wada M, Shimizu T, et al. 2012;
in fetal and postnatal hearts. J Anat 187 in skeletal defects. Curr Stem Cell Res Ther 8 Creation of human cardiac cell sheets using
(3): 641647. (3): 243252. pluripotent stem cells. Biochem Biophys Res
Brown JS, Shaw RJ. 2010; Reconstruction of Hare JM, Fishman JE, Gerstenblith G, et al. Commun 425(2): 321327.
the maxilla and midface: introducing a 2012; Comparison of allogeneic vs autolo- Mayhew TM, Gregson C, Pharaoh A, et al.
new classication. Lancet Oncol 11(10): gous bone marrow-derived mesenchymal 1998; Numbers of nuclei in different tissue
10011008. stem cells delivered by transendocardial compartments of fetal ventricular myocar-
Cannon TY, Strub GM, Yawn RJ, et al. 2012; injection in patients with ischemic dium from 16 to 35 weeks of gestation.
Oromandibular reconstruction. Clin Anat cardiomyopathy: the POSEIDON random- Virchows Arch 433(2): 167172.
25(1): 108119. ized trial. J Am Med Assoc 308(22): Mayhew TM, Pharaoh A, Austin A, et al.
Chen KG, Mallon BS, McKay RD, et al. 2014; 23692379. 1997; Stereological estimates of nuclear
Human pluripotent stem cell culture: con- Huttenbach Y, Ostrowski ML, Thaller D, et al. number in human ventricular
siderations for maintenance, expansion, 2001; Cell proliferation in the growing cardiomyocytes before and after birth ob-
and therapeutics. Cell Stem Cell 14(1): human heart: MIB-1 immunostaining in tained using physical disectors. J Anat
1326. preterm and term infants at autopsy. 191(1): 107115.
Choi BR, Salama G. 2000; Simultaneous Cardiovasc Pathol 10(3): 119123. Mohan AT, Al-Ajam Y, Mosahebi A. 2013;
maps of optical action potentials and cal- Langer R, Vacanti JP. 1993; Tissue engineer- Trends in tertiary breast reconstruction:
cium transients in guinea-pig hearts: ing. Science 260(5110): 920926. literature review and single centre experi-
mechanisms underlying concordant Makkar RR, Smith RR, Cheng K, et al. 2012; ence. Breast 22(2): 173178.
alternans. J Physiol 529(1): 171188. Intracoronary cardiosphere-derived cells for Nemeno-Guanzon JG, Lee S, Berg JR, et al.
Chugh AR, Beache GM, Loughran JH, et al. heart regeneration after myocardial 2012; Trends in tissue engineering for
2012; Administration of cardiac stem cells infarction (CADUCEUS): a prospective, blood vessels. J Biomed Biotechnol 2012
in patients with ischemic cardiomyopathy: randomised phase 1 trial. Lancet 379 (2012); 956345.
the SCIPIO trial: surgical aspects and in- (9819): 895904. Okano T, Yamada N, Sakai H, et al. 1993;
terim analysis of myocardial function and Masumoto H, Ikuno T, Takeda M, et al. A novel recovery system for cultured
viability by magnetic resonance. Circula- 2014; Human iPS cell-engineered cardiac cells using plasma-treated polystyrene
tion 126(11, suppl 1): S5464. tissue sheets with cardiomyocytes and dishes grafted with poly(N-

Copyright 2015 John Wiley & Sons, Ltd. J Tissue Eng Regen Med (2015)
DOI: 10.1002/term
H. Komae et al.

isopropylacrylamide). J Biomed Mater Sako Y. 1971d; [Electron microscopic studies vascularized myocardial tissues. FASEB J
Res 27(10): 12431251. on the development of the human embry- 20(6): 708710.
Sakaguchi K, Shimizu T, Horaguchi S, onic myocardium. 4. Cell junction]. Jpn Shimizu T, Yamato M, Isoi Y, et al. 2002;
et al. 2013; In vitro engineering of Circ J 35(11): 15041508. Fabrication of pulsatile cardiac tissue
vascularized tissue surrogates. Sci Rep Sekine H, Shimizu T, Sakaguchi K, et al. grafts using a novel three-dimensional cell
3: 1316. 2013; In vitro fabrication of functional sheet manipulation technique and
Sako Y. 1971a; [Electron microscopic studies three-dimensional tissues with temperature-responsive cell culture sur-
on the development of the human embry- perfusable blood vessels. Nat Commun faces. Circ Res 90(3): e40.
onic myocardium. 1. Myobrils]. Jpn Circ 4: 1399. Tuan RS, Chen AF, Klatt BA. 2013; Cartilage
J 35(11): 14911496. Shimizu T, Sekine H, Isoi Y, et al. 2006a; regeneration. J Am Acad Orthop Surg 21
Sako Y. 1971b; [Electron microscopic studies Long-term survival and growth of (5): 303311.
on the development of the human embry- pulsatile myocardial tissue grafts Wong CH, Wei FC. 2010; Microsurgical free
onic myocardium. 2. Mitochondria and engineered by the layering of cardio- ap in head and neck reconstruction. Head
glycogen]. Jpn Circ J 35(11): 14971500. myocyte sheets. Tissue Eng 12(3): Neck 32(9): 12361245.
Sako Y. 1971c; [Electron microscopic studies 499507. Yildirimer L, Thanh NT, Seifalian AM. 2012;
on the development of the human embry- Shimizu T, Sekine H, Yang J, et al. 2006b; Skin regeneration scaffolds: a multimodal
onic myocardium. 3. Granules]. Jpn Circ J Polysurgery of cell sheet grafts overcomes bottom-up approach. Trends Biotechnol 30
35(11): 15011503. diffusion limits to produce thick, (12): 638648.

Supporting information on the internet


The following supporting information may be found in the online version of this article:

Figure S1. HiPSCMs cultured in vitro and in vivo: electron microscopy images of hiPSCMs cultured for 1 month in vitro
(A) and in vivo (B). Myobrils (arrows) were being fused and elongated in both images, but the density of sarcomere
units in the hiPSCMs cultured in vivo was obviously higher; intracellular structures of hiPSCMs cultured in vivo were
also more developed
Video S1. Optical mapping of electric membrane potential; the electric conduction ows in a non-random pattern (400
consecutive frames, 0.001 s/frame)
Video S2. Three-layer hiPSCM sheets transplanted after 6 months; wholly synchronized regular beating can be seen in
the transplanted area

Copyright 2015 John Wiley & Sons, Ltd. J Tissue Eng Regen Med (2015)
DOI: 10.1002/term

You might also like