You are on page 1of 13

N. Bursac, M. Papadaki, R. J. Cohen, F. J. Schoen, S. R. Eisenberg, R. Carrier, G.

Vunjak-Novakovic and L. E. Freed


Am J Physiol Heart Circ Physiol 277:433-444, 1999.

You might find this additional information useful...

This article cites 41 articles, 18 of which you can access free at:
http://ajpheart.physiology.org/cgi/content/full/277/2/H433#BIBL

This article has been cited by 13 other HighWire hosted articles, the first 5 are:
Region of slowed conduction acts as core for spiral wave reentry in cardiac cell monolayers
J. W. Lin, L. Garber, Y. R. Qi, M. G. Chang, J. Cysyk and L. Tung
Am J Physiol Heart Circ Physiol, January 1, 2008; 294 (1): H58-H65.
[Abstract] [Full Text] [PDF]

Biomimetic approach to cardiac tissue engineering


M Radisic, H Park, S Gerecht, C Cannizzaro, R Langer and G Vunjak-Novakovic
Phil Trans R Soc B, August 29, 2007; 362 (1484): 1357-1368.
[Abstract] [Full Text] [PDF]

In vitro tissue engineering of a cardiac graft using a degradable scaffold with an extracellular
matrix-like topography

Downloaded from ajpheart.physiology.org on May 24, 2010


O. Ishii, M. Shin, T. Sueda and J. P. Vacanti
J. Thorac. Cardiovasc. Surg., November 1, 2005; 130 (5): 1358-1363.
[Abstract] [Full Text] [PDF]

Mathematical model of oxygen distribution in engineered cardiac tissue with parallel channel
array perfused with culture medium containing oxygen carriers
M. Radisic, W. Deen, R. Langer and G. Vunjak-Novakovic
Am J Physiol Heart Circ Physiol, March 1, 2005; 288 (3): H1278-H1289.
[Abstract] [Full Text] [PDF]

Medium perfusion enables engineering of compact and contractile cardiac tissue


M. Radisic, L. Yang, J. Boublik, R. J. Cohen, R. Langer, L. E. Freed and G. Vunjak-Novakovic
Am J Physiol Heart Circ Physiol, February 1, 2004; 286 (2): H507-H516.
[Abstract] [Full Text] [PDF]

Medline items on this article's topics can be found at http://highwire.stanford.edu/lists/artbytopic.dtl


on the following topics:
Physiology .. Cardiac Muscle
Physiology .. Heart Muscle
Engineering .. Biomedical Engineering
Physiology .. Rats
Updated information and services including high-resolution figures, can be found at:
http://ajpheart.physiology.org/cgi/content/full/277/2/H433

Additional material and information about AJP - Heart and Circulatory Physiology can be found at:
http://www.the-aps.org/publications/ajpheart

This information is current as of May 24, 2010 .

AJP - Heart and Circulatory Physiology publishes original investigations on the physiology of the heart, blood vessels, and
lymphatics, including experimental and theoretical studies of cardiovascular function at all levels of organization ranging from the
intact animal to the cellular, subcellular, and molecular levels. It is published 12 times a year (monthly) by the American Physiological
Society, 9650 Rockville Pike, Bethesda MD 20814-3991. Copyright © 2005 by the American Physiological Society. ISSN: 0363-6135,
ESSN: 1522-1539. Visit our website at http://www.the-aps.org/.
Cardiac muscle tissue engineering: toward an
in vitro model for electrophysiological studies
N. BURSAC,1,2 M. PAPADAKI,1 R. J. COHEN,1 F. J. SCHOEN,3 S. R. EISENBERG,2
R. CARRIER,1 G. VUNJAK-NOVAKOVIC,1 AND L. E. FREED1
1Division of Health Sciences and Technology, Massachusetts Institute of Technology,

Cambridge 02139; 2Department of Biomedical Engineering, Boston University, Boston 02215; and
3Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts 02115

Bursac, N., M. Papadaki, R. J. Cohen, F. J. Schoen, physiological behavior of normal and pathological car-
S. R. Eisenberg, R. Carrier, G. Vunjak-Novakovic, and diac tissues. Such studies are currently performed in
L. E. Freed. Cardiac muscle tissue engineering: toward an in one-dimensional cardiac strands and two-dimensional
vitro model for electrophysiological studies. Am. J. Physiol. (2-D) isotropic, anisotropic, and photolithographically
277 (Heart Circ. Physiol. 46): H433–H444, 1999.—The objec-
patterned monolayers using optical mapping tech-
tive of this study was to establish a three-dimensional (3-D)
in vitro model system of cardiac muscle for electrophysiologi- niques (9, 10, 27). Impulse propagation studies cannot
cal studies. Primary neonatal rat ventricular cells containing be performed in 3-D myocyte aggregates (17, 30) be-
lower or higher fractions of cardiac myocytes were cultured on cause of their small size (100–300 µm) and isopotential
polymeric scaffolds in bioreactors to form regular or enriched nature. Other 3-D cultures of cardiac myocytes grown
cardiac muscle constructs, respectively. After 1 wk, all con- on microcarrier beads (1, 31), collagen fibers (1), syn-

Downloaded from ajpheart.physiology.org on May 24, 2010


structs contained a peripheral tissue-like region (50–70 µm thetic, biodegradable polymeric templates (3, 12), or in
thick) in which differentiated cardiac myocytes were orga- collagen gels (8) have not yet been evaluated electro-
nized in multiple layers in a 3-D configuration. Indexes of cell physiologically.
size (protein/DNA) and metabolic activity (tetrazolium conver- The goal of the present work was to establish a 3-D in
sion/DNA) were similar for constructs and neonatal rat
vitro model system for impulse propagation studies in
ventricles. Electrophysiological studies conducted using a
linear array of extracellular electrodes showed that the
cardiac muscle using tissue engineering principles.
peripheral region of constructs exhibited relatively homoge- This approach relies on the use of primary cells in
neous electrical properties and sustained macroscopically conjunction with biodegradable polymer scaffolds (13,
continuous impulse propagation on a centimeter-size scale. 18) and tissue culture bioreactors (11, 12). The polymer
Electrophysiological properties of enriched constructs were scaffold provides a 3-D substrate for cell attachment
superior to those of regular constructs but inferior to those of and tissue formation, whereas the mixing of culture
native ventricles. These results demonstrate that 3-D cardiac medium in the bioreactor promotes mass transfer of
muscle constructs can be engineered with cardiac-specific nutrients and gases to the forming tissue. Primary
structural and electrophysiological properties and used for in neonatal rat ventricular cells were cultured on polymer
vitro impulse propagation studies. scaffolds in bioreactors to form tissue constructs, which
myocyte; impulse propagation; electrophysiology; three- were characterized histologically, biochemically, and
dimensional electrophysiologically and compared with neonatal and
adult rat ventricular tissues.

MATERIALS AND METHODS


CULTURED CARDIAC MYOCYTES offer many advantages for
developmental, physiological, and pharmacological stud- All experiments involving animals were performed accord-
ies of cardiac tissue because they allow for direct cell ing to the Institutional Committee on Animal Care of the
manipulation and control of environmental parameters Massachusetts Institute of Technology, which follows federal
without interference from the compensatory feedback and state guidelines.
mechanisms that exist in vivo. Compared with mono- Cardiac myocyte preparation. Primary cultures of cardiac
layer cultures, it has been suggested that three- myocytes were prepared by enzymatic digestion of ventricles
obtained from neonatal (2 day old) Sprague-Dawley rats
dimensional (3-D) multilayered cultures of cardiac myo-
(Taconic), as previously described (44). Briefly, ventricles (n ⫽
cytes more closely resemble intact cardiac tissue with 50, 5 litters in 3 independent studies) were incubated with
respect to cellular differentiation (8) and electrical 0.1% trypsin overnight and dissociated in four to five sequen-
properties (38, 39). Three-dimensional cardiac myocyte tial steps using 0.1% collagenase. Isolated cells were resus-
cultures could thus be used for in vitro studies of pended in culture medium [DMEM, supplemented with 10%
cardiac tissue development and function and, if suffi- fetal bovine serum (FBS), 50 U/ml penicillin and 10 mM
ciently large and functional, for in vivo cardiac repair. HEPES, all obtained from GIBCO-BRL].
Impulse propagation studies in cultures of cardiac Two experimental groups were established as follows (Fig.
myocytes can improve our understanding of the electro- 1A): 1) a regular group of ventricular cells isolated as
described above and 2) an enriched group with a higher
fraction of cardiac myocytes, prepared from the regular group
The costs of publication of this article were defrayed in part by the by centrifugation at 600 rpm for 5 min, followed by two
payment of page charges. The article must therefore be hereby preplatings, 75 min each (Fig. 1A); cells that remained
marked ‘‘advertisement’’ in accordance with 18 U.S.C. Section 1734 unattached after the second preplating were used. Cell yields
solely to indicate this fact. were ⬃6 ⫻ 106 and 5 ⫻ 106 cells/ventricle for the regular and

0363-6135/99 $5.00 Copyright r 1999 the American Physiological Society H433


H434 CARDIAC MUSCLE TISSUE ENGINEERING

Downloaded from ajpheart.physiology.org on May 24, 2010

Fig. 1. A: model system for tissue engineering. Cells from neonatal rat ventricles were seeded onto polymer
scaffolds and cultured for 7 days to form regular and cardiac myocyte-enriched constructs. B: electrophysiological
setup. Tissue constructs were studied using an extracellular microelectrode array (inset) under controlled
environmental conditions in a 37°C/5% CO2 perfused chamber. Stimulation was bipolar, and extracellular
recordings were unipolar with reference to a Ag-AgCl electrode placed 3.5 cm away from the microelectrode array.
CARDIAC MUSCLE TISSUE ENGINEERING H435

enriched group, respectively. Cell viability was 91 ⫾ 3%, as incubated with avidin-biotin complex reagent and 3,38-
assessed by trypan blue exclusion. diaminobenzidine (Sigma). Ten randomly selected fields (0.3 ⫻
Monolayer studies. Cells from the regular and enriched 0.4 mm2 each) from six coverslips from each group were
groups were cultured in monolayers at a cell density of 1.3 ⫻ analyzed using videomicroscopy and NIH Image 1.60 soft-
104 cells/cm2 in 12-well dishes, T75 flasks, and on glass ware to estimate cardiac myocyte fraction as a percentage of
coverslips to assess spontaneous contractions and biochemi- cell area stained positively for tropomyosin.
cal and immunohistochemical parameters, respectively. After Ventricles and 7-day constructs were fixed in 2% glutaralde-
2 days of static culture, monolayers were placed on an orbital hyde for 10 min, rinsed in PBS, and immersed in 10% neutral
shaker set to 75 rpm. Medium was completely replaced on day buffered Formalin (Sigma). Samples were embedded in paraf-
3 and by 50% on day 5. Spontaneous contractions were fin, sectioned at 5 µm, and stained with hematoxylin and
assessed by videomicroscopy, by manually counting the num- eosin (H ⫹ E) for general evaluation and Masson’s trichrome
ber of beats per minute using five randomly selected fields stain for collagen assessment. Immunohistochemical stain-
(0.3 ⫻ 0.4 mm2 each) per plate and six plates per experimen- ing for tropomyosin was used to assess the fraction of cardiac
tal group, on days 3, 5, and 7. Cells in T75 flasks were myocytes in constructs. Sections were incubated with 1 mg/ml
removed after 7 days by a 5-min incubation with 0.05% trypsin (Sigma) at 37°C for 15 min and 0.3% hydrogen
trypsin-EDTA (GIBCO-BRL) and counted, and a suspension peroxide for 30 min, blocked with horse serum for 30 min, and
of 2 ⫻ 106 cells/ml was stored at ⫺20°C for determination of incubated with antisarcomeric tropomyosin as described
DNA and protein contents and lactate dehydrogenase (LDH) above. A humidified chamber was used for all incubation
activity per cell. Cells on glass coverslips were fixed with steps. Sections were counterstained with Mayer’s hematoxy-
HistoCHOICE (Amresco) for immunohistochemical analysis. lin (Sigma) and coverslipped using glycerol mounting media
3-D tissue culture studies. Cells from the regular and (Sigma). Specificity of staining for tropomyosin was con-
enriched groups were cultured on polyglycolic acid (PGA) firmed by staining for sarcomeric ␣-actin, another myocyte-

Downloaded from ajpheart.physiology.org on May 24, 2010


scaffolds, which are highly porous (97%) meshes of randomly specific protein, using otherwise identical methodology. Con-
entangled 13-µm fibers formed as 5 ⫻ 2-mm (diameter ⫻
struct macroscopic architecture was assessed from stained
thickness) disks (Fig. 1A; Ref. 13). Briefly, scaffolds were
tissue sections using videomicroscopy and NIH Image 1.60
prewetted in culture medium, positioned on thin stainless
software.
steel wires using segments of silicone tubing, and fixed to a
Transmission electron microscopy. Samples were fixed in
silicone stopper placed in the mouth of a spinner flask (8
Karnovsky’s reagent (0.1 M sodium cacodylate with 2%
scaffolds per flask) (12). Flasks were filled with 120 ml of
paraformaldehyde and 2.5% glutaraldehyde, pH ⫽ 7.4), post-
culture medium, placed in a humidified 37°C, 5% CO2 incuba-
fixed in 2% osmium tetroxide, dehydrated in ethanol in
tor with the side arm caps loosened to permit gas exchange,
propylene oxide, and embedded in Poly/Bed812 (Poly-
and mixed at 50 rpm using a magnetic stir bar. After 24 h,
sciences). Sections were cut at 60 nm, stained with lead
flasks were inoculated with cells (8 ⫻ 106 cells per scaffold).
citrate and uranyl acetate, and examined using a transmis-
Culture medium was replaced by 100% on day 3 and by 50%
sion electron microscope (JEOL-100CX, JEOL).
on day 5. Cell-polymer constructs (n ⫽ 22, from 3 independent
studies) were harvested after 7 days for morphometric, Media analysis. Physiological ranges of PO2 (115–130
histological, biochemical, and electrophysiological assess- mmHg), PCO2 (48–55 mmHg), and pH (7.21–7.33) were
ments. maintained for the duration of cultivation, as measured by a
Ventricular tissues. To verify the analytical methods, evalu- blood gas analyzer (IL 1610, Instrumentation Laboratory).
ate the developmental state of cardiac myocytes in constructs, Glucose and lactate concentrations were measured using a
and establish baseline values for parameters studied in glucose/lactate analyzer (2300 StatPlus, YSI). The activity of
engineered constructs that were not readily found in the LDH in the culture media was monitored using a LDH-L
literature, two control groups were examined. Adult ven- reagent kit (Chiron Diagnostics). Media samples were soni-
tricles (n ⫽ 10) were obtained from 3- to 4-mo-old Sprague- cated using a Sonic Dismembrator (Vibra-Cell, Sonics and
Dawley rats following anesthesia by intramuscular injection Materials), and absorbance was measured at 340 nm (Spec-
of 65 mg/kg ketamine and 5 mg/kg xylazine (Sigma). Hearts tronic 1001⫹, Milton Roy) against cell-free medium. An LDH
were rapidly removed, and ventricular sections were excised activity of 1 U/l corresponded to 3,600 cells in monolayers.
from 1 mm below the atrioventricular groove to 1–2 mm DNA and protein assays. DNA and protein assays were
above the apex. For electrophysiological studies, full-thick- performed on engineered constructs and native ventricles
ness pieces of the ventricular wall (⬃9 ⫻ 7 mm2, 2–4 mm using modifications of previously described methods (7).
thick) were then prepared by making two longitudinal cuts Samples were homogenized in buffer (1 N ammonium hydrox-
parallel to the base-apex line. Neonatal ventricles (n ⫽ 10, ide/2% Triton X-100, 0.04 ml/mg wet wt) for 1 min. For the
from 3 litters) were obtained from 2-day-old rats following DNA assay, homogenates were incubated at 37°C for 10 min,
decapitation. For electrophysiological studies, full-thickness diluted with assay buffer (100 mM NaCl, 1 mM EDTA, 10 mM
pieces of the ventricular wall (⬃6 ⫻ 4 mm2, 1.5–2.5 mm thick) Tris, pH 7.00), and centrifuged. DNA contents of superna-
were prepared by bisecting the ventricle. Smaller pieces of tants were determined using a spectrofluorometer (PTI) and
the adult and neonatal ventricles (7–13 mg wet wt) were used calf thymus DNA as a standard (7). DNA contents measured
for biochemical and histological assessments. The properties for regular and enriched monolayers were comparable (7.1 ⫾
of neonatal and adult ventricles were compared with those of 0.2 pg/cell) and consistent with published values (7).
constructs without a priori assumption that the engineered For protein assays, the viscosity of homogenates was
tissue resembled either of the native ventricular tissues. reduced by several passages through a 26-gauge needle. After
Histological and immunohistochemical assessments. Cells centrifugation, protein concentration was measured in the
on glass coverslips were incubated for 30 min with mouse supernatant using a Bio-Rad DC protein assay kit and a
antisarcomeric tropomyosin monoclonal antibody (clone CH1, microplate spectrophotometer (MR5000, Dynatech). Regular
Sigma) diluted 1:100 in PBS containing 0.5% Tween 20 and and enriched monolayers had comparable protein contents
1.5% horse serum and then for 30 min with a secondary (290 pg/cell), resulting in protein-to-DNA ratios of 41 mg/mg
antibody (Vectastain), diluted 1:200. Coverslips were then that were consistent with published values (29).
H436 CARDIAC MUSCLE TISSUE ENGINEERING

Metabolic activity assays. Metabolic activities of cells within applied at a rate of 60 beats/min, starting at a pacing voltage
constructs and ventricular tissues were assessed by the of 0.1 V, which was then increased in 0.1-V increments until
uptake and enzymatic reduction of the tetrazolium dye the sample was captured (i.e., until each pacing impulse was
3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide followed by a recorded tissue response). The corresponding
(MTT; Sigma). Samples (2–15 mg wet wt) were rinsed with pacing voltage, defined as the excitation threshold, repre-
PBS and incubated with MEM (GIBCO-BRL) without phenol sented the lowest stimulus that produced a stable propaga-
red and 0.5 mg/ml MTT for 4 h on an orbital shaker at 37°C tion (for at least 1 min at a rate of 60 beats/min) over the
and 60 rpm. Medium was replaced with an equal volume of length of the recording array. For the next 20–30 min, the
0.1 N HCl in absolute isopropanol and pipetted directly sample was continuously paced at 60 beats/min using pacing
through the constructs to solubilize the resulting formazan amplitudes 1.5 times higher than the excitation threshold,
crystals. After 10 min of incubation at 37°C, the absorbance and responses were recorded every 4–5 min for a period of 1
was read at 570 nm, using a microplate spectrophotometer. min. The pacing rate was then increased every 5 min by 30
Electrophysiological assessment. An electrophysiological beats/min, and responses at each rate were recorded for the
system was custom-designed to enable stimulation and record- last 40 s, similar to the protocol in Ref. 37. The maximum
ing of unipolar extracellular potentials in constructs and pacing frequency at which the sample could be captured for at
ventricular tissues under controlled environmental condi- least 5 min was defined as the maximum capture rate. After
tions using a linear array of microelectrodes (Fig. 1B). A reaching the maximum capture rate, stimulation was stopped
cylindrical Plexiglas chamber was tightly fitted inside an for 10 min and then reapplied at 30 and 60 beats/min for 5
electrically grounded brass casing placed on a 37°C heater min each to check for reproducibility of the recorded wave-
(VWR). The brass case distributed the heat evenly through forms. At the end of the experiment, double and triple
the chamber and served as an electrostatic shield. The extrastimuli and rapid stimulation at frequencies above the
chamber was gassed with a prewarmed mixture of 5% CO2 in maximum capture rate were applied in an attempt to induce

Downloaded from ajpheart.physiology.org on May 24, 2010


air and filled with 50 ml of culture medium (DMEM with 15 arrhythmia.
mM HEPES, 4.5 g/l glucose), which was recirculated (at 60 All recorded signals were amplified and band-pass filtered
ml/min for constructs and 120 ml/min for ventricular tissues) between 0.3 and 1,000 Hz. The unfiltered noise level was 35
using a pulseless gear pump (Cole-Parmer). Temperature and µV, peak to peak, with virtually no 60-Hz component. Analog
pH were maintained at 37 ⫾ 0.1°C and 7.32 ⫾ 0.02, respec- recordings were digitized at a sampling rate of 3 kHz using a
tively. 16-bit analog-to-digital board (AT-MIO-16X, National Instru-
A photomicrograph of the microelectrode array is shown in ments), real-time displayed using LabView data acquisition
Fig. 1B. All microelectrodes were made of insulated tungsten software, and stored and analyzed using MATLAB (The
wire and had uninsulated tips with diameters of 50 ⫾ 6 µm Mathworks).
(Microprobe). Two electrodes for bipolar stimulation were Activation times at each recording electrode were deter-
positioned 200 µm apart and connected to a programmable mined as the minima of five-point derivatives (2) of the
cardiac stimulator (SEC-3102, Nihon Kohden). Eight record- low-pass filtered signals. The stimulus-activation time inter-
ing electrodes were positioned 500 µm apart in a linear array, vals at each electrode (conduction times) were plotted against
1.5 to 5 mm from the stimulating site. Exact distances the corresponding distances and fitted by linear regression.
between electrodes were measured using a microscope and The conduction velocity of a propagated beat was calculated
NIH 1.60 image analysis software. Shielded cables connected as the inverse slope of the best linear fit (16). The peak-to-
recording electrodes to bioelectric amplifiers (AB.601G, Ni- peak (p-p) amplitudes of the responses were determined from
hon Kohden). A reference Ag-AgCl electrode (WPI) was placed linearly detrended signals around the activation times. Re-
in the medium 3.5 cm away from the microelectrode array. cording sites with very low or fractionated (polyphasic)
Samples were placed in a tissue holder 2–3 mm under the activity were ignored.
surface of the culture medium, secured using Teflon screws, For each tissue sample, p-p amplitudes at each electrode
and left to equilibrate for 15 min. An XYZ mechanical and conduction velocities were averaged from recordings
micropositioner (Taurusr, WPI) was used to gradually ad- made during the initial 20 min of pacing at 60 beats/min (i.e.,
vance the microelectrode array toward either the top surface over at least 200 beats). Conduction velocity, maximum
of the construct or the epicardial surface of the ventricle, and amplitude, and average amplitude were calculated, respec-
pacing impulses were simultaneously applied (3–5 V, 1-ms tively, as the averages of conduction velocities, maximum p-p
pulses at a rate of 60 beats/min). The position of the array was amplitudes, and all p-p amplitudes from all samples within a
fixed at the point where the amplitudes of the recorded group. The maximum and average amplitudes, respectively,
responses appeared maximal, and a recording protocol was represented local and spatially averaged properties of con-
performed as follows. structs or ventricles.
Spontaneous beating, if present, was recorded for 3–5 min. Statistics. Data were calculated as means ⫾ SE and
After 15 min, monophasic pacing pulses (1-ms duration) were analyzed using either a paired t-test or one-way ANOVA

Table 1. Morphometric and biochemical parameters in 7-day constructs


Construct Weight and Dimensions Cell Number Cell Number per Glucose Consumption Lactate Production
in Construct, Construct Lost During Rate per Construct, Rate per Construct,
Group Wet weight, mg Thickness, mm Circular area, mm2 ⫻106 Cultivation, ⫻106 g · l⫺1 · day⫺1 g · l⫺1 · day⫺1

Regular 35.45 ⫾ 3.33 1.34 ⫾ 0.11 20.14 ⫾ 0.82 3.75 ⫾ 0.59 5.36 ⫾ 0.19 2.21 ⫾ 0.02 1.43 ⫾ 0.12
(20.95 ⫾ 0.65)
Enriched 31.08 ⫾ 1.71 1.30 ⫾ 0.07 22.92 ⫾ 0.58* 3.00 ⫾ 0.46 4.63 ⫾ 0.31 5.37 ⫾ 0.95* 2.51 ⫾ 0.11*
(17.68 ⫾ 1.12)
Values are means ⫾ SE; n ⫽ 5 constructs. Parenthetical values are cumulative lactate dehydrogenase activity per construct, in U/l. Regular
and enriched groups were prepared as defined in MATERIALS AND METHODS. * Statistically significant difference between regular and enriched
groups.
CARDIAC MUSCLE TISSUE ENGINEERING H437

Fig. 2. Histology and immunohistochem-


istry of enriched constructs and native
ventricles. A: construct periphery (hema-
toxylin and eosin, H ⫹ E). B: construct
center (H ⫹ E). C: construct periphery
(tropomyosin). D: construct periphery
(tropomyosin). E: neonatal ventricle
(tropomyosin). F: adult ventricle (tropo-
myosin). A–C: magnification, ⫻400; bar,

Downloaded from ajpheart.physiology.org on May 24, 2010


50 µm. D–F: magnification, ⫻1,000; bar,
20 µm. Brown color indicates tropomyosin-
positive cells. Asterisks denote polymer
fibers; arrows point to cross striations.

followed by Fisher’s protected least significant difference post layers in the enriched than in the regular group (7 ⫾ 1
hoc test. To determine time-dependence trends for beating vs. 5 ⫾ 1 layers, respectively). Cells in this outermost
rates in monolayer cultures, a univariate repeated-measures zone formed a continuous, 3-D tissuelike structure by
ANOVA was used. Differences were considered statistically
attaching to other cells, spreading along the randomly
significant when P ⬍ 0.05. All calculations were performed
using SuperANOVA III for Macintosh. oriented PGA fibers, and forming bridges between the
fibers (Fig. 2, A and C). Distinct cardiac bundles,
RESULTS spatially oriented groups of cells (⬎100 µm in size), and
interstitial collagen septa were not observed. Ran-
Monolayer cultures. After 24 h of culture, cardiac
myocytes from both the regular and enriched groups domly oriented cells in the peripheral zone exhibited a
started to contract spontaneously and by day 3–4 variety of shapes, from elongated cells spread on the
formed synchronously contracting networks. Rates of polymer fibers to round unattached cells, as assessed
contraction decreased significantly between culture histologically. The majority of the cells expressed the
days 3 and 7 (P ⬍ 0.05) in monolayers from both groups. muscle-specific proteins sarcomeric tropomyosin (Fig.
At day 7, enriched monolayers had significantly higher 2, C and D) and sarcomeric ␣-actin (data not shown).
cardiac myocyte fractions and contraction rates than Immediately below the peripheral zone was a 60- to
regular monolayers (60.5 ⫾ 1.5 vs. 43.8 ⫾ 0.5% of the 70-µm-thick region consisting mainly of cells that did
culture area, P ⬍ 0.04, and 169 ⫾ 8 vs. 132 ⫾ 10 not express tropomyosin. At the construct center, cells
beats/min, P ⬍ 0.01), which is consistent with previous were sparsely distributed and either elongated, express-
reports (22). ing tropomyosin, or round, with pyknotic nuclei and
Construct morphology. After 7 days of culture, cell- acidophilic cytoplasm (Fig. 2B).
polymer constructs appeared discoid [⬃5 ⫻ 1.3 mm Cross striations were present in cells in the periph-
(diameter ⫻ thickness); Table 1]. The peripheral zone eral zone of the constructs as well as in neonatal and
was 50–70 µm thick (Fig. 2A) and consisted of more cell adult ventricles, as assessed immunohistochemically
H438 CARDIAC MUSCLE TISSUE ENGINEERING

Fig. 3. Transmission electron micro-


graph of an enriched construct. A: well-
organized myofilaments (Mfl) with
clearly defined sarcomeres, z-lines (Z),
and abundant glycogen granules (Gly).
Magnification, ⫻26,000; bar, 1 µm. B:
several mitochondria (Mito) located be-
tween myofilaments. Magnification,
⫻36,000; bar, 400 nm. C: desmosomes
(Des) at lateral border of adjacent car-
diac myocytes. Magnification, ⫻18,000;
bar, 250 nm. D: intercalated disk (ID)

Downloaded from ajpheart.physiology.org on May 24, 2010


with visible desmosomes. Magnifica-
tion, ⫻18,000; bar, 250 nm.

(Fig. 2, D–F). The presence of subcellular elements construct groups (1.00 ⫾ 0.20 and 1.30 ⫾ 0.11, respec-
characteristic of cardiac myocytes, including myofila- tively).
ments with well-defined sarcomeres, z-lines, glycogen Ventricular tissues from neonatal and adult rats had
granules (Fig. 3A), and mitochondria (Fig. 3B) in the respectively six- and threefold higher DNA contents per
outermost layer of constructs, was demonstrated by unit wet weight (an index of cellularity) than engi-
transmission electron microscopy (TEM). Cell-to-cell neered constructs from either group (P ⬍ 0.01, Fig. 4A),
connections were demonstrated by the presence of which is consistent with the relatively acellular appear-
desmosomes (Fig. 3C) and intercalated disks (Fig. 3D). ance of the construct centers (Fig. 2B). Relative cell
Construct composition. After 3 days, the respective size, assessed from the ratio of total protein to DNA,
numbers of viable cells present in enriched and regular was comparable for cells in constructs, neonatal ven-
constructs were 66 and 57% of those seeded at time 0, as tricles, and monolayers and lower than for cells in adult
calculated from medium LDH levels. LDH release be- ventricles (P ⬍ 0.01) (Fig. 4B). This finding was consis-
tween culture days 3 and 7 was one-third of that tent with the relative cross-sectional areas of cells in
between days 0 and 3, indicating that the cell death constructs and neonatal and adult ventricles observed
rate decreased with cultivation time. At 7 days, cell histologically (Fig. 2, D–F, respectively). The MTT
numbers in enriched and regular constructs were 38 conversion per unit DNA (an index of metabolic activ-
and 47% of the respective numbers seeded at time 0, as ity) was similar for constructs and neonatal ventricles
determined by the DNA content of constructs. For and was slightly higher in adult ventricles (Fig. 4C).
comparison, 7-day cell monolayers from both groups Construct electrophysiology. Spontaneous, macro-
contained 61 ⫾ 6% of the initially plated cells. The scopic contractions of engineered constructs were visu-
number of cells seeded at time 0 (8 million per PGA ally observed in flasks between days 2 and 4 of cultiva-
disk) could be accounted for by summing cell numbers tion, which indicated the presence of intercellular
in constructs at day 7 (determined from DNA content) communication. At day 7 the majority of constructs and
and in the medium over 7 days (calculated from cumu- native ventricles exhibited transient spontaneous beat-
lative LDH activity/construct) (Table 1), implying that ing lasting for 1–10 contractions (Fig. 5A), which may
no significant cell proliferation occurred during the have resulted from reentrant or triggered activity (4).
cultivation period. Glucose consumption and lactate Electrical stimulation resulted in impulse propagation
production rates were higher in enriched than in in the peripheral cardiac tissue-like zone of the con-
regular constructs (P ⬍ 0.005, Table 1), whereas the structs. In contrast, impulses failed to propagate when
lactate-to-glucose molar ratios were similar for both the electrodes were advanced toward the central acellu-
CARDIAC MUSCLE TISSUE ENGINEERING H439

cally excitable for up to 4 wk of culture (data not


shown).
Representative examples of impulse propagation in
an enriched construct, a neonatal ventricle, and an
adult ventricle are shown in Fig. 6, A–C, respectively.
Propagating extracellular waveforms in constructs and
native tissues showed fairly smooth, biphasic shapes
with distinct downward deflections that enabled confi-
dent determination of activation times and implied
nondecremental, macroscopically continuous propaga-
tion without wave collisions (34). Notches in extracellu-
lar waveforms occasionally observed in constructs (see
E3 in Fig. 6A) may have reflected asynchronous excita-
tion in adjacent groups of cells caused by the presence
of empty space, polymer fibers, and/or necrotic tissue
(36). Conduction times in ventricles and constructs
increased linearly with distance over 5 mm (Fig. 6D,

Downloaded from ajpheart.physiology.org on May 24, 2010


Fig. 4. Cellularity, hypertrophy, and metabolic activity indexes of
constructs and ventricles. A: DNA content per unit wet weight (ww).
B: protein per unit DNA. C: MTT conversion per unit DNA. Data
represent means ⫾ SE of 5 samples. * Statistical difference between
constructs and native ventricles; † statistical difference between
neonatal and adult ventricles.

lar region of the constructs. All 7-day constructs were


electrically excitable and could be captured over a wide
range of pacing frequencies (up to 270 beats/min, Fig. 5,
B-D). Step increases in construct pacing frequency
resulted in transient decreases in conduction velocity to Fig. 5. Electrophysiological recordings. A: short transient spontane-
steady-state values (data not shown). Rapid stimula- ous beating at a rate of 70 beats/min, which lost regularity after 6 s.
tion induced short tachyarrhythmias with rates close to B, C, and D: steady-state responses after ⱖ4 min of pacing at rates of
the maximum capture rates in 3 of 6 enriched con- 80, 150, and 200 beats/min, respectively. E: short, 5-s tachyarrhyth-
mia at ⬃190 beats/min, apparently induced by 4 rapid stimuli at 250
structs, 2 of 6 regular constructs (Fig. 5E), 1 of 10 adult beats/min. S and R indicate the stimulus spike and the construct
ventricles, and 0 of 10 neonatal ventricles. A separate response, respectively. All tracings were recorded from enriched
experiment showed that constructs remained electri- constructs using the same electrode.
Downloaded from ajpheart.physiology.org on May 24, 2010
CARDIAC MUSCLE TISSUE ENGINEERING
H440
CARDIAC MUSCLE TISSUE ENGINEERING H441

Table 2. Electrophysiological parameters in 7-day constructs and native ventricles


Excitation Conduction Maximum Average Maximum Capture
Group n Threshold, V Velocity, cm/s Amplitude, mV Amplitude, mV Rate, beats/min

Constructs
Regular* 6 2.70 ⫾ 0.24 9.35 ⫾ 0.27 0.52 ⫾ 0.05 0.26 ⫾ 0.09 111.7 ⫾ 9.5
Enriched* 6 2.97 ⫾ 0.30 11.89 ⫾ 0.46† 0.90 ⫾ 0.14† 0.43 ⫾ 0.14 175.0 ⫾ 21.3†
Ventricles
Neonatal 10 0.74 ⫾ 0.20 21.82 ⫾ 1.48 31.91 ⫾ 3.53 18.34 ⫾ 4.31 475.0 ⫾ 25.0
Adult 10 1.34 ⫾ 0.17‡ 31.69 ⫾ 4.44‡ 25.82 ⫾ 2.81 14.62 ⫾ 3.59 281.2 ⫾ 21.0‡
Data represent means ⫾ SE; n ⫽ no. of constructs or ventricles. * Significant difference between constructs and ventricles; † significant
difference between enriched and regular constructs; ‡ significant difference between neonatal and adult ventricles.

regression coefficients ⬎ 0.98), implying similar conduc- eral cardiac tissue-like zone in which differentiated
tion velocities between adjacent electrodes and thus cardiac myocytes were organized in multiple layers and
relatively homogeneous electrical properties through- attached to other cells and/or polymer fibers in a 3-D
out the cardiac tissue-like zone. configuration. Impulse propagation studies carried out
Conduction velocities descended in the following using an array of extracellular microelectrodes demon-
order: adult ventricles, neonatal ventricles, enriched strated that the peripheral cardiac tissue-like zone of
constructs, and regular constructs (Table 2, Fig. 6D) constructs sustained macroscopically continuous im-
(P ⬍ 0.001). Lower conduction velocities measured in

Downloaded from ajpheart.physiology.org on May 24, 2010


pulse propagation (Fig. 6A) that depended on the
neonatal than in adult ventricles were consistent with fraction of seeded cardiac myocytes (Table 2). Func-
previously published values (40, 41). Conduction veloci- tional constructs may thus enable in vitro electrophysi-
ties measured in enriched constructs were ⬃30 and ological studies that may complement those currently
50% as high as those in adult and neonatal ventricles, carried out using thin ventricular slices (5, 14, 35) and
respectively (P ⬍ 0.001) and were 27% higher than in monolayers of cardiac myocytes (9).
regular constructs (P ⬍ 0.02). Excitation thresholds Structurally, constructs were 5 ⫻ 1.3-mm (diameter ⫻
were higher in engineered constructs than in native thickness) disks and contained a 50- to 70-µm-thick
ventricles (P ⬍ 0.001, Table 2) and were lower in outer cardiac tissue-like zone composed of cells that
neonatal than in adult ventricles (P ⬍ 0.01). Excitation expressed sarcomeric tropomyosin (Fig. 2C) and con-
thresholds of constructs and ventricles thus varied tained myofilaments, desmosomes, and intercalated
inversely with cellularity indexes (Table 2, Fig. 4A). disks (Fig. 3D). For comparison, a recently reported (8)
Maximum and average amplitudes were signifi- heart muscle model system based on cardiac myocyte-
cantly lower in constructs than in native ventricles populated 3-D collagen gels (15 mm long ⫻ 8 mm
(P ⬍ 0.0001, Table 2). Maximum amplitudes were wide ⫻ 180 µm thick) contained several layers of
1.7-fold higher in enriched than in regular constructs differentiated cells at the edges and less concentrated
(P ⬍ 0.002), whereas average amplitudes did not differ cells centrally. The small thickness of the cardiac
significantly. The range of recorded amplitudes was tissue-like zone in constructs (Fig. 2A) and collagen
⬃3-fold higher in constructs than in native ventricles gels (8) can be attributed to the low survival rate of
(data not shown). Maximum capture rates differed metabolically demanding cardiac myocytes located more
significantly (P ⬍ 0.001) among groups and descended than 50 µm from a source of gas exchange (15).
in the following order: neonatal ventricles, adult ven- The molar ratios of lactate to glucose of 1.0–1.3
tricles, enriched constructs, and regular constructs indicated aerobic cell metabolism in the constructs (21).
(Table 2). The higher maximum capture rates in neona- Compared with the regular group, enriched constructs
tal ventricles than in adult ventricles were consistent had higher glucose consumption rates (Table 1), prob-
with the higher resting heart rates and higher toler- ably due to the relatively higher fraction of myocytes.
ance to ischemia previously reported for neonatal ven- The absence of cell proliferation in constructs (Table 1)
tricles (47). was consistent with the previous findings that neonatal
DISCUSSION ventricular cardiac myocytes lose their ability to prolif-
erate after 2–3 days in vitro (45), whereas fibroblasts
The present study demonstrates that 3-D cardiac proliferate slowly in 3-D cultures (20).
muscle constructs with cardiac-specific structural and Electrophysiologically, impulse propagation in con-
electrophysiological properties can be engineered in structs was studied on a macroscopic level using a
vitro using isolated cells and biodegradable polymer linear array of extracellular electrodes (Fig. 1B). Inter-
scaffolds. In particular, constructs contained a periph- electrode distances of 500 µm were selected on the basis

Fig. 6. Impulse propagation (representative 66 ms) in a construct from the enriched group (A), neonatal ventricle
(B), and adult ventricle (C). The extracellular waveform is shown propagating from the electrode closest to the
stimulus site (E1) toward the furthest electrode still in the sample (E6). Simultaneous deflections at the beginning
of traces (S) represent stimulus artifacts. Amplitude ranges for each electrode were adjusted to best display
recorded response waveforms. D: plot of conduction times vs. distances relative to E1, which was assigned the
coordinates (0,0). Conduction velocities were calculated as inverse slopes of best fit lines.
H442 CARDIAC MUSCLE TISSUE ENGINEERING

of previously reported in vivo and ex vivo epicardial cardiac myocytes due to injury during cultivation.
mapping studies (6, 46, 48). Bipolar point stimulation However, it is more likely that the cultivation of cardiac
and unipolar recording (16, 25) in the custom-designed myocytes on 3-D biomaterial scaffolds in tissue culture
test chamber (Fig. 1B) did not adversely affect samples bioreactors (Fig. 1A) promoted differentiated cellular
with respect to their electrical properties (waveform phenotype and function. In support of this hypothesis,
shapes were stable) or structure (no apparent tissue Sperelakis (38) showed that 3-D aggregates composed
damage was observed histologically). Automated data of electrically differentiated cardiac myocytes did not
analysis was facilitated by the high average signal-to- contract spontaneously but responded to electrical
noise ratios (of ⬃10 and 470 for constructs and native stimulation.
ventricles, respectively). Whereas 1- to 5-V amplitude, The aim of the present study was to demonstrate
1-ms duration electrical pulses were sufficient to induce basic cardiac-specific features in constructs and to
impulse propagation in slices of ventricles and in the evaluate construct structure and electrophysiological
peripheral zone of 7-day constructs, it was difficult to properties on a macroscopic (tissue) level, rather than
overdrive 7-day confluent monolayers of neonatal car- on a cellular level. In ongoing work, we are expanding
diac myocytes even when using stimuli of twice this our electrophysiological studies to include whole cell
amplitude and duration. In addition, impulse propaga- clamp and sharp microelectrode intracellular record-
tion in monolayers could not be assessed using extracel- ings and assessment of the spatial distribution of the
lular electrodes because of fractionation and low ampli- gap junctional protein connexin 43 (23). We are also
tudes of recorded waveforms. These findings may be attempting to culture constructs with a thicker cardiac
due to 3-D electrotonic interactions between cells (9) tissue-like zone by direct perfusion of constructs during

Downloaded from ajpheart.physiology.org on May 24, 2010


and relatively high cell density around the stimulating cultivation (to improve mass transfer) and by cocultur-
and recording electrodes in 3-D constructs compared ing cardiac myocytes with microvascular endothelial
with 2-D monolayers. cells (as a first step toward inducing vascularization).
The inferior electrophysiological properties of con- In conclusion, cardiac-specific features of engineered
structs compared with native ventricles (Table 2) can cardiac muscle constructs were demonstrated structur-
be attributed to differences in their macroscopic tissue ally and electrophysiologically and were related to the
architecture. In particular, the relatively high excita- cellular composition of constructs. The 3-D multilayer
tion thresholds (24) and low response amplitudes were structure in conjunction with macroscopic impulse
associated with low construct cellularity (Fig. 4A). Low propagation in engineered constructs can offer advan-
maximum capture rates and conduction velocities in tages for in vitro studies of cardiac muscle. In addition,
constructs probably resulted from decreased cell cou- structurally and functionally improved 3-D engineered
pling, the presence of intercellular clefts, and geometric cardiac muscle constructs could be eventually applied
current-to-load mismatches (due to tissue discontinui- in vivo. To date, attempts to regenerate cardiac tissue
ties) (9, 26). Other mechanisms that could contribute to have involved the injection of different muscle cell
inferior construct electrophysiological properties in- types (33, 43) or small tissue fragments (19) into the
clude cell depolarization, reduced excitability, and heart. Implantation of cardiac muscle constructs with a
slower repolarization resulting from injury during isola- defined shape instead of isolated cells could potentially
tion and/or cultivation (32, 39). Intracellular recordings improve the efficiency and localization of tissue repair.
would be necessary to test the proposed mechanisms.
Compared with enriched constructs, lower conduc- N. Bursac and M. Papadaki contributed equally to this study.
tion velocities, maximum capture rates, and ampli- We thank R. Langer for advice, R. Padera for help with animal
surgery, H. Shing for carrying out the transmission electron micros-
tudes in regular constructs probably resulted from 1) copy, Y. Lee for help establishing the electrophysiological recording
the higher fraction of noncardiomyocytic cells, which system, and J. Merok, H. Cho, and P. Gupta for help with biochemical
would be expected to form high-resistance junctions assays.
with cardiac myocytes (28) and act as passive current This work was supported by National Aeronautics and Space
sinks (9), and 2) the thinner cardiac tissue-like zone Administration Grant NAG9-836.
Address for reprint requests and other correspondence: L. E.
(Table 1). Lower maximum capture rates in the regular Freed, Massachusetts Institute of Technology, Div. of Health Science
than enriched constructs could also be due to the and Technology, MIT, Bldg. E25–342, Cambridge, MA 02139 (E-mail:
relatively longer duration of cellular action potentials lfreed@mit.edu).
(as previously observed in fibrotic compared with nor- Received 7 October 1998; accepted in final form 8 March 1999.
mal cardiac tissue; Ref. 42).
Neonatal and adult ventricular tissues did not ex- REFERENCES
hibit spontaneous beating ex vivo in a previous (39) or
1. Akins, R. E., N. A. Schroedl, S. R. Gonda, and C. R. Hartzell.
the present study. In contrast, enzymatically isolated Neonatal rat heart cells cultured in simulated microgravity. In
ventricular cardiac myocytes cultured in monolayers Vitro Cell Dev. Biol. 33: 337–343, 1997.
are known to revert to a less differentiated phenotype, 2. Blanchard, S., W. Smith, R. Damiano, D. Molter, R. Ideker,
depolarize, and regain spontaneous contractile activity and J. Lowe. Four digital algorithms for activation detection
for as yet unknown reasons (39). In the present study, from unipolar epicardial electrograms. IEEE Trans. Biomed.
Eng. 36: 256–261, 1989.
visible spontaneous contractions in constructs ceased 3. Carrier, R., M. Papadaki, M. Rupnick, F. J. Schoen, N.
after 4 days of cultivation. This finding might be Bursac, R. Langer, L. E. Freed, and G. Vunjak-Novakovic.
attributed to gradual depolarization and decoupling of Cardiac tissue engineering: cell seeding, cultivation parameters
CARDIAC MUSCLE TISSUE ENGINEERING H443

and tissue construct characterization. Biotechnol Bioeng. In 25. Rogers, J., P. Bayly, R. Ideker, and W. Smith. Quantitative
press. techniques for analyzing high-resolution cardiac mapping data.
4. Cranefield, P., and R. Aronson. Cardiac Arrhythmias: The IEEE Eng. Med. Biol. Mag. 17: 62–72, 1998.
Role of Triggered Activity and Other Mechanisms. Mount Kisco, 26. Rohr, S., and B. Salzberg. Characterization of impulse propa-
NY: Futura, 1988. gation at the microscopic level across geometrically defined
5. Davidenko, J., R. Salomonsz, A. Pertsov, W. Baxter, and J. expansions of excitable tissue: multiple site optical recording of
Jalife. Effects of pacing on stationary reentrant activity: theoreti- transmembrane voltage (MSORTV) in patterned growth heart
cal and experimental study. Circ. Res. 77: 1166–1179, 1995. cell cultures. J. Gen. Physiol. 104: 287–309, 1994.
6. De Bakker, J., F. van Cappele, and M. Janse. Slow conduc- 27. Rohr, S., and B. Salzberg. Multiple site optical recording of
tion in the infarcted human heart: ‘‘zigzag’’ course of activation. transmembrane voltage (MSORTV) in patterned growth heart
Circulation 88: 915–926, 1993. cell cultures: assessing electrical behavior, with microsecond
7. Downs, T., and M. Wilfinger. Fluorometric quantification of resolution, on a cellular and subcellular scale. Biophys. J. 67:
DNA in cells and tissue. Anal. Biochem. 131: 538–547, 1983. 1301–1315, 1994.
8. Eschenhagen, T., C. Fink, U. Remmers, H. Scholz, J. 28. Rook, M., A. Van Ginneken, B. De Jonge, A. El Aoumari, D.
Wattchow, J. Weil, W. Zimmerman, H. Dohmen, H. Schafer, Gros, and H. Jongsma. Differences in gap junction channels
between cardiac myocytes, fibroblasts, and heterologous pairs.
N. Bishopric, T. Wakatsuki, and E. Elson. Three-dimensional
Am. J. Physiol. 263 (Cell Physiol. 32): C959–C977, 1992.
reconstitution of embryonic cardiomyocytes in a collagen matrix:
29. Schroedl, N., and C. Hartzell. Myocytes and fibroblasts exhibit
a new heart muscle model system. FASEB J. 11: 683–694, 1997.
functional synergism in mixed cultures of neonatal rat heart
9. Fast, V., B. Darrow, J. Saffitz, and A. Kleber. Anisotropic
cells. J. Cell. Physiol. 117: 326–332, 1983.
activation spread in heart cell monolayers assessed by high-
30. Schubert, B., R. Bodewei, S. Hering, and A. Wollenberger.
resolution optical mapping. Role of tissue discontinuities. Circ.
Cell-attached patch clamp measurement of macroscopic rapid
Res. 79: 115–127, 1996. inward sodium current in cultured heart cell reaggregates. J.
10. Fast, V., and A. Kleber. Microscopic conduction in cultured Mol. Cell. Cardiol. 19: 1129–1139, 1987.

Downloaded from ajpheart.physiology.org on May 24, 2010


cardiac strands of neonatal rat heart cells measured with 31. Shahar, A., S. Reuveny, Y. David, C. Budu, and A. Shain-
voltage-sensitive dyes. Circ. Res. 73: 914–925, 1993. burg. Cerebral neurons, skeletal myoblasts, and cardiac muscle
11. Freed, L. E., and G. Vunjak-Novakovic. Culture of organized cells cultured on macroporous beads. Biotechnol. Bioeng. 43:
cell communities. Adv. Drug Delivery Res. 33: 15–30, 1998. 826–831, 1994.
12. Freed, L., and G. Vunjak-Novakovic. Microgravity tissue 32. Shaw, R., and Y. Rudy. Electrophysiological effects of acute
engineering. In Vitro Cell Dev. Biol. 33: 381–385, 1997. myocardial ischemia: a theoretical model of altered excitability
13. Freed, L. E., G. Vunjak-Novakovic, R. Biron, D. Eagles, D. and action potential duration. Cardiovasc. Res. 35: 256–272,
Lesnoy, S. Barlow, and R. Langer. Biodegradable polymer 1997.
scaffolds for tissue engineering. Biotechnology 12: 689–693, 33. Soonpa, M., A. Daud, G. Koh, M. Klug, K. Kim, H. Wang, and
1994. L. Field. Potential approaches for myocardial regeneration.
14. Girouard, S., K. Laurita, and D. Rosenbaum. Unique proper- Ann. NY Acad. Sci. 752: 446–454, 1995.
ties of cardiac action potentials recorded with voltage-sensitive 34. Spach, M., R. Bar, and G. Serwer. Collision of excitation waves
dyes. J. Cardiovasc. Electrophysiol. 7: 1024–1037, 1996. in the dog Purkinje system. Circ. Res. 29: 499–511, 1971.
15. Guyton, A. Transport of oxygen and carbon dioxide in the blood 35. Spach, M., P. Dolber, and J. Heidlage. Properties of discontinu-
and body fluids. In: Textbook of Medical Physiology, edited by A. ous anisotropic propagation at a microscopic level. Ann. NY
Guyton. Philadelphia, PA: Saunders, 1986, p. 493–503. Acad. Sci. 591: 62–74, 1990.
16. Knisley, S., and B. Hill. Effects of bipolar point and line 36. Spach, M., P. Dolber, and J. Heidlage. Resolution of discontinu-
stimulation in anisotropic rabbit epicardium: assessment of the ous versus continuous propagation: microscopic mapping of the
critical radius of curvature for longitudinal block. IEEE Trans. derivatives of extracellular potential waveforms. In: Cardiac
Biomed. Eng. 42: 957–966, 1995. Electrophysiology: From Cell to Bedside, edited by D. Zipes and J.
17. Kunysz, A., A. Shrier, and L. Glass. Overdrive suppression of Jalife. Philadelphia, PA: Saunders, 1990, p. 139–148.
spontaneously beating chick heart cell aggregates: experiment 37. Spach, M., J. Kootsey, and J. Sloan. Active modulation of
and theory. Am. J. Physiol. 269 (Heart Circ. Physiol. 38): H1153– electrical coupling between cardiac cells of the dog: a mechanism
H1164, 1995. for transient and steady state variations in conduction velocity.
18. Langer, R., and J. Vacanti. Tissue engineering. Science 260: Circ. Res. 51: 347–362, 1982.
920–926, 1993. 38. Sperelakis, N. Cultured heart cell reaggregate model for study-
19. Leor, J., M. Patterson, M. Quinones, L. Kedes, and R. ing cardiac toxicology. Environ. Health Perspect. 26: 243–267,
Kloner. Transplantation of fetal myocardial tissue into the 1978.
infarcted myocardium of rat. Circulation 94: II-322–II-336, 1996. 39. Sperelakis, N., and G. Haddad. Developmental changes in
20. Mauch, C., A. Hatamochi, K. Scharffetter, and T. Krieg. membrane electrical properties of the heart. In: Physiology and
Pathophysiology of the Heart (3rd ed.), edited by N. Sperelakis.
Regulation of collagen synthesis in fibroblasts within a three-
Norwell, MA: Kluwer Academic, 1995, p. 669–700.
dimensional collagen gel. Exp. Cell Res. 178: 493–503, 1988.
40. Sun, L., M. Legato, T. Rosen, S. Steinberg, and M. Rosen.
21. Opie, L. Fuels: aerobic and anaerobic metabolism. In: The Heart:
Sympathetic innervation modulates ventricular impulse propaga-
Physiology, from Cell to Circulation (3rd ed.), edited by L. Opie.
tion and repolarisation in immature rat heart. Cardiovasc. Res.
Philadelphia, PA: Lippincott-Raven, 1998, p. 295–342. 27: 459–463, 1993.
22. Orita, H., M. Fukasawa, S. Hirooka, H. Uchino, K. Fukui, 41. Suzuki, J., H. Tsubone, and S. Sugano. Characteristics of
and M. Washio. Modulation of cardiac myocyte beating rate and ventricular activation and recovery patterns in the rat. J. Vet.
hypertrophy by cardiac fibroblasts isolated from neonatal rat Med. Sci. 54: 711–716, 1992.
ventricle. Jpn. Circ. J. 57: 912–920, 1993. 42. Swynghedauw, B., B. Chevalier, D. Charlemagne, P. Man-
23. Papadaki, M., N. Bursac, R. Langer, F. Schoen, G. Vunjak- sier, and F. Carre. Cardiac hypertrophy, arrhythmogenicity and
Novakovic, and L.E. Freed. Towards a functional engineered the new myocardial phenotype. II. The cellular adaptational
cardiac muscle: effects of cell culture substrate and medium process. Cardiovasc. Res. 35: 6–12, 1997.
serum concentration. In: Society for Biomaterials, 25th Annual 43. Taylor, D., B. Atkins, P. Hungspreugs, T. Jones, M. Reedy,
Meeting Transactions. Minneapolis, MN: Soc. Biomaterials, 1999, K. Hutcheson, D. Glower, and W. Kraus. Regenerating func-
p. 359. tional myocardium: improved performance after skeletal myo-
24. Ramza, B., R. Tan, T. Osaka, and R. Joyner. Cellular mecha- blast transplantation. Nat. Med. 4: 929–933, 1998.
nism of the functional refractory period in ventricular muscle. 44. Toraason, M., M. Luken, M. Breitenstein, J. Krueger, and
Circ. Res. 66: 147–162, 1990. R. Biagini. Comparative toxicity of allylamine and acrolein in
H444 CARDIAC MUSCLE TISSUE ENGINEERING

cultured myocytes and fibroblasts from neonatal rat heart. 47. Xie, L., M. Takano, and A. Noma. Development of inwardly
Toxicology 56: 107–113, 1989. rectifying K⫹ channel family in rat ventricular myocytes. Am. J.
45. Undo, H., M. B. Perryman, R. Roberts, and M. D. Schneider. Physiol. 272 (Heart Circ. Physiol. 41): H1741–H1750, 1997.
Differentiation of cardiac myocytes after mitogen withdrawal
exhibits three sequential states of the ventricular growth re- 48. Yin, H., N. El-Sherif, E. B. Caref, G. Ndrepepa, R. Levin,
sponse. J. Cell Biol. 107: 1911–1918, 1988. N. Isber, K. Stergiopolus, M. Assadi, W. Gough, and M.
46. Wit, A., S. Dillon, J. Coromilas, A. Saltman, and B. Wal- Restivo. Actions of lidocaine on reentrant ventricular rhythms
decker. Anisotropic reentry in the epicardial border zone of in the subacute myocardial infarction period in dogs. Am. J.
myocardial infarcts. Ann. NY Acad. Sci. 591: 86–108, 1990. Physiol. 272 (Heart Circ. Physiol. 41): H299–H309, 1997.

Downloaded from ajpheart.physiology.org on May 24, 2010

You might also like