You are on page 1of 16

THE

JOURNAL • RESEARCH • www.fasebj.org

Cardiomyocyte–myofibroblast contact dynamism


is modulated by connexin-43
Francisca Schultz,*,1,2 Pamela Swiatlowska,*,1 Anita Alvarez-Laviada,*,1 Jose L. Sanchez-Alonso,*
Qianqian Song,* Antoine A. F. de Vries,† Daniël A. Pijnappels,† Emily Ongstad,‡ Vania M. M. Braga,§
Emilia Entcheva,{ Robert G. Gourdie,‡ Michele Miragoli,k,#,3 and Julia Gorelik*,4
*National Heart and Lung Institute, Imperial College London, London, United Kingdom; †Department of Cardiology, Leiden University
Medical Center, Leiden, The Netherlands; ‡Center for Heart and Regenerative Medicine, Virginia Tech Carilion Research Institute, Roanoke,
Virginia, USA; §Department of Respiratory Sciences, Faculty of Medicine, National Heart and Lung Institute, Imperial College London,
London, United Kingdom; {Department of Biomedical Engineering, George Washington University, Washington, DC, USA; kHumanitas
Clinical and Research Center, Milan, Italy; and #Department of Medicine and Surgery, University of Parma, Parma, Italy

ABSTRACT: Healthy cardiomyocytes are electrically coupled at the intercalated discs by gap junctions. In infarcted
hearts, adverse gap-junctional remodeling occurs in the border zone, where cardiomyocytes are chemically and
electrically influenced by myofibroblasts. The physical movement of these contacts remains unquantified. Using
scanning ion conductance microscopy, we show that intercellular contacts between cardiomyocytes and myofi-
broblasts are highly dynamic, mainly owing to the edge dynamics (lamellipodia) of the myofibroblasts. Decreasing
the amount of functional connexin-43 (Cx43) at the membrane through Cx43 silencing, suppression of Cx43 traf-
ficking, or hypoxia-induced Cx43 internalization attenuates heterocellular contact dynamism. However, we found
decreased dynamism and stabilized membrane contacts when cellular coupling was strengthened using 4-
phenylbutyrate (4PB). Fluorescent-dye transfer between cells showed that the extent of functional coupling be-
tween the 2 cell types correlated with contact dynamism. Intercellular calcein transfer from myofibroblasts to
cardiomyocytes is reduced after myofibroblast-specific Cx43 down-regulation. Conversely, 4PB-treated myofi-
broblasts increased their functional coupling to cardiomyocytes. Consistent with lamellipodia-mediated contacts,
latrunculin-B decreases dynamism, lowers physical communication between heterocellular pairs, and reduces Cx43
intensity in contact regions. Our data show that heterocellular cardiomyocyte–myofibroblast contacts exhibit high
dynamism. Therefore, Cx43 is a potential target for prevention of aberrant cardiomyocyte coupling and myofi-
broblast proliferation in the infarct border zone.—Schultz, F., Swiatlowska, P., Alvarez-Laviada, A., Sanchez-Alonso,
J. L., Song, Q., de Vries, A. A. F., Pijnappels, D. A., Ongstad, E., Braga, V. M. M., Entcheva, E., Gourdie, R. G., Miragoli,
M., Gorelik, J. Cardiomyocyte–myofibroblast contact dynamism is modulated by connexin-43. FASEB J.
33, 000–000 (2019). www.fasebj.org
KEY WORDS: heart failure • myocardial infarction • Cx43 • heterocellular coupling

ABBREVIATIONS: 4PB, 4-phenylbutyrate; CM–CM, cardiomyocyte– Communication between cells is necessary to maintain
cardiomyocyte; CM–MFB, cardiomyocyte–myofibroblast; Cx43, connexin- proper organ development and function. Coupling between
43; KD, knockdown; MFB–MFB, myofibroblast–myofibroblast; shRNA, both cells of the same cell type as well as between different cell
short hairpin RNA; siRNA, small interfering RNA; SICM, scanning ion
conductance microscopy; a-SMA, a-smooth muscle actin types (heterocellular coupling) occurs ubiquitously through-
1
These authors contributed equally to this work.
out numerous tissues (1–5). Formation of cell–cell contacts
2
Current affiliation: Springer Nature, London, United Kingdom. via stable junctions is the basis of cellular crosstalk. Twelve
3
Correspondence: Department of Medicine and Surgery, University of connexin-hemichannel proteins form a stable gap junction
Parma, Via Gramsci 14, 43124 Parma, Italy. E-mail: michele.miragoli@
unipr.it
between 2 cells. This coupling has been shown to be present in
4
Correspondence: National Heart and Lung Institute, 4th Floor, Imperial different cell types, such as neurons, vascular smooth muscle
Centre for Translational and Experimental Medicine, Imperial College cells, and cardiomyocytes (1–7). Independently of the tissue
London, Hammersmith Campus, Du Cane Rd., London W12 0NN,
United Kingdom. E-mail: j.gorelik@imperial.ac.uk and composition of different connexin subunits, these gap
junctions perform the same electrical and chemical functions.
doi: 10.1096/fj.201802740RR
This article includes supplemental data. Please visit http://www.fasebj.org to Intercellular communication controls critical physio-
obtain this information. logic processes, facilitates dynamic tissue responses, and

0892-6638/19/0033-0001 © FASEB 1
g by Saab Medical Library, American Univ of Beruit (UAB) Levant U (193.188.128.21) on June 30, 2019. The FASEB Journal Vol. ${article.issue.getVolume()}, No. ${article.issue.get
promotes tissue remodeling triggered by pathologic cardiomyocytes undergo structural remodeling, with Cx43
events. In the healthy heart, operating at the highest level moving from the intercalated discs at the polar ends of the
of multicellular synchronicity of action, cellular packing cell to the lateral surfaces. This lateralization of Cx43 in-
and strong electrical communication are essential for its creases the likelihood of cardiomyocyte-to-myofibroblast
operation. Interdispersed among the highly coupled adult (CM–MFB) coupling and the associated risk of ar-
ventricular cardiomyocytes are a large number of fibro- rhythmia (21).
blasts and myofibroblasts (8, 9). These mesenchymal cells Myofibroblasts appear to have complex roles, as,
are 2 to 3 times more abundant than cardiomyocytes, but depending on the setting, they have been implicated in
because of their smaller size, they only comprise ;20% of both pro- or antiarrhythmic modulation. Heterocellular
the cardiac muscle tissue (8). Interactions between car- electrotonic communication has been shown to be det-
diomyocytes and fibroblasts occur in the heart at different rimental to cardiac function in computer-modeling
levels. The paracrine functions of fibroblasts have been and experimental studies because myofibroblasts
characterized, including secretion of TGF-b and other are thought to directly depolarize cardiomyocytes,
soluble factors (10, 11). Of particular note, fibroblasts slowing action potential conduction and promoting
synthesize extracellular matrix fibers, such as collagen and ectopic electrical activity (14, 23–26). On the other hand,
fibronectin, which form the 3-dimensional scaffold of the myofibroblast–cardiomyocyte coupling by gap junctions
heart that supports the cardiomyocytes. It has also been can have a positive impact on conduction across discon-
shown that cardiomyocytes and fibroblasts in the heart are tinuities in networks of cardiomyocytes (15). In a study
mechanically coupled either via the extracellular matrix or of recipient and donor atria, where a scar formed in the
via adherens junctions; however, little is known about the region of the suture, evidence of successful restoration
importance of such interactions (12). Finally, electrical of electrical activity was found (27). By acting as passive
coupling may occur between the 2 cell types (9), although electrical conduits for current flow, myofibroblasts were
to what extent this occurs and what the role of hetero- thus shown to contribute to the electrical synchronization
cellular coupling in the normal heart is remains poorly between host and graft tissue following heart trans-
understood. In contrast to cardiomyocytes, fibroblasts are plantation. Similarly, expression of exogenous Cx43 in
motile cells that constantly form new contacts with infarct scar myofibroblasts has been demonstrated to
neighboring cells and that remodel the extracellular ma- improve conduction and stabilize the electrical rhythm in
trix; both of these processes depend on the actin cytoskel- preclinical animal models of cardiac injury (28). In addi-
eton motility of cardiac fibroblasts. The potential impact of tion to the modulation of electrical properties, there is
the motility of fibroblasts on the coupling between car- growing evidence that heterocellular mechanical interac-
diomyocytes and fibroblasts has not yet been investigated. tions have a pivotal role in the structure and function of
After an ischemic event, such as myocardial infarction, the CM–MFB coupling zone and scar (29, 30).
myofibroblasts proliferate, specifically in the region in Therefore, myofibroblasts appear to have complex
which the injury occurred, as well as in the surrounding regulatory roles; depending on the setting, they are im-
area. Fibroblasts are replaced by, or differentiate into, plicated in either proarrhythmic or antiarrhythmic mod-
myofibroblasts, which are characterized by a number of ulation of the heart after a pathologic event. Nevertheless,
distinct markers, including de novo expression of a-smooth the detailed cellular and molecular mechanisms by which
muscle actin (a-SMA) (13). Myofibroblasts have been im- physical heterocellular contacts mediate effects in cardiac
plicated in the formation of myocardial scars and scar disease remain poorly characterized. In this study, we
expansion, which in turn has been linked to an increase in developed an in vitro model of heterocellular contacts that
arrhythmogenesis (14–18). A narrow atypical zone of are formed during heart failure and we investigated
myocardial tissue that surrounds the scar is where close whether electrical and mechanical coupling are indepen-
contacts between myofibroblasts and cardiomyocytes oc- dent processes or whether they act in synergy during
cur; this is in contrast to the scar proper, where few car- CM–MFB zone expansion. We show that dynamism of
diomyocytes survive because of necrosis (19). interactions with this model depends on gap-junctional
Transformation or infiltration of myofibroblasts is Cx43 levels.
thought to be triggered by a number of signals, includ-
ing local inflammatory reactions, mechanical stress, and
various cytokines and biochemical factors (9, 17). Cul- MATERIALS AND METHODS
tured myofibroblasts have been shown to couple to car-
diomyocytes through gap junction-based interactions (14). Neonatal rat cells used for contact motility studies
However, it is not clear whether gap-junctional coupling
occurs between myofibroblasts and cardiomyocytes in Isolation of neonatal rat ventricular cardiomyocytes and myofi-
the diseased myocardium in vivo, although a recent study broblasts from 1–2-d-old Sprague-Dawley rats was done in ac-
has shown that myofibroblasts can electrically couple to cordance with the guidelines of the Home Office Animal
cardiomyocytes in the infarct border zone of isolated, (Scientific Procedures) Act of 1986 of the United Kingdom. Car-
Langendorff-perfused mouse hearts ex vivo (20). Camelliti diomyocytes and myofibroblasts were isolated and cultured as
previously described (31). Monocultures of cardiomyocytes or
et al. (21) demonstrated that the main connexin sub- myofibroblasts were seeded into MatTek dishes (13 cm2; MatTek,
types that are expressed by resident fibroblasts in infarct Ashland, MA, USA) or plastic-bottom dishes (35 mm, CytoOne;
scars are connexin-43(Cx43) and connexin-45 (Cx45) (14, USA Scientific, Ocala, FL, USA). For the coculture experiments,
21, 22). It is thought that when an ischemic event occurs, cardiomyocytes were also plated in dishes, with myofibroblasts

2 Vol. 33 September 2019 The FASEB Journal x www.fasebj.org SCHULTZ ET AL.


g by Saab Medical Library, American Univ of Beruit (UAB) Levant U (193.188.128.21) on June 30, 2019. The FASEB Journal Vol. ${article.issue.getVolume()}, No. ${article.issue.get
seeded on top on the subsequent day. The myofibroblasts were differences in speed between the start and end. Oxygen levels
detached as previously described (31), spun, and resuspended in were measured at the end of the session and were found to not
1 ml HBSS per 1 million cells. The myofibroblasts were then have returned to normoxia before the end of the set of scans.
labeled by incubating for 20 min at 37°C and 1% CO2 with either Cocultures were treated with latrunculin-B (100 mM), at 37°C for
Vybrant DiI (excitation/emission: 549/565 nm; Thermo Fisher 24 h in order to disrupt actin stress fibers (41).
Scientific, Waltham, MA, USA) or WGA-488 (wheat germ
agglutinin-488, excitation/emission: 495/519 nm; Thermo Fisher
Scientific) for recognition afterward. The myofibroblasts were Scanning ion conductance microscopy
then seeded in with the cardiomyocytes and were left to make
gap junctions for 24 h before imaging. Scanning ion conductance microscopy (SICM) was used to
image the contact area between cells (Fig. 1A). In brief, SICM
provides continuous topographical images in hopping mode via
Adult rat fibroblasts used for contact a feedback-controlled nanopipette scan with minimal spatial drift
motility studies (a few nm) (23). SICM is a noncontact method that allows the
imaging of the topography of intact cells (23, 42, 43). Surface
Studies involving adult rats were performed in accordance with topographical images of cardiomyocytes and myofibroblasts
the Guide for the Care and Use of Laboratory Animals [National were acquired by SICM at 25°C in a low-calcium physiologic
Institutes of Health (NIH), Bethesda, MD, USA]. For the myo- solution (144 mM NaCl (MilliporeSigma, Burlington, MA, USA),
cardial infarction model, male adult Sprague-Dawley rats 5 mM KCl (MilliporeSigma), 1 mM MgCl2 (MilliporeSigma),
(250–300 g) were anesthetized and underwent proximal coro- 10 mM HEPES, in 2 l dH2O, pH 7.4) or in HBSS, in order to avoid
nary ligation to induce chronic myocardial infarction. Rats contraction. A large scan (60–100 3 60–100 mm) was carried out
were allowed to recover for 16 wk to induce heart failure. to identify the contact areas between cells. After that, a smaller,
Time-matched control animals that did not undergo surgery higher resolution scan of ;15 3 15 mm of the contact area was
were used as control. After 16 wk, in vivo pressure–volume carried out and repeated for 1 h (loop) to visualize changes in the
analysis was performed before the hearts were explanted, contact area between 2 cells (Fig. 1A, C). Afterward, the images
weighed, and prepared for cell isolation as previously described (15 3 15 mm) were processed in chronological order using
(32–34). Fibroblasts were collected from the supernatant of the the CellTrack software (44) (http://bio.cse.ohio-state.edu/CellTrack/;
cardiomyocyte isolation. Cardiomyocytes were spun at 400 g for Fig. 1A, C). The program returns the average of the speed (pixel
1 min, after which the supernatant containing the fibroblasts was per frame) from the randomly selected points within the selected
collected. This supernatant was then spun at 1000 g for 10 min to contact area (Fig. 1C). By knowing the time of each acquired
pellet the fibroblasts. The supernatant was removed, and the image (hh:mm:ss) and the number of pixels for each image
pellet was resuspended in DMEM containing 10% fetal bovine (512 3 512 pixels), it was possible to evaluate the average
serum, 1% antibiotic/antimycotic solution, and 1% L-glutamine. movement velocity (mm/min) of a selected area.
Myofibroblasts were plated in 25-cm2 flasks and grown until
confluency.
SICM in combination with confocal microscopy

Modulation of gap junctions and Cx43 Surface topographical images of cardiomyocytes and myofibro-
blasts were acquired by SICM at 25°C in low-calcium buffer. For
the visualization of Cx43 at the junctions, cells were incubated for
Various methods to modulate gap junctions were used. Over-
24 h with the first-generation adenovirus vector encoding
expression of Cx43 fused at its C terminus to enhanced green-
Cx43–GFP (Adeno-X Expression System; Takara, Kyoto, Japan)
fluorescent protein (eGFP) was achieved using a first-generation
at a multiplicity of infection of 9. Cx43–GFP was excited at 473 nm
adenovirus vector (Cx43–eGFP) and knockdown (KD) was car-
with a Stradus 473 laser (Vortran, Sacramento, CA, USA) and
ried out using small interfering RNA (siRNA) (199927; Thermo
confocal images were made at 3100 magnification using a Pho-
Fisher Scientific, Waltham, MA, USA) or vesicular stomatitis vi-
tomultiplier Detection System (PTI). Using the SICM setup, the
rus G-protein-pseudotyped lentiviral vectors encoding GFP
tip of the pipette was aligned with the laser beam. The surface of
and a Cx43-specific short hairpin RNA (shRNA) (35, 36). Cx43
the cell was then scanned as a conventional SCIM image. The
modulation by zonula occludens-1 (ZO-1) was inhibited by a
same area was subsequently scanned with the laser to visualize
membrane-permeable peptide inhibitor that contains the Cx43
the Cx43. The resulting images could be overlapped with the
C-terminal postsynaptic density-95/disks-large/ZO-1 (PDZ)-
corresponding topography image of the surface (Fig. 1B).
binding domain (aCT1 peptide) (37). Cardiomyocytes and
myofibroblasts were incubated, separately or in cocultures, for
;4 h with the aCT1 peptide (100 mM). Increase in cellular
Immunofluorescence microscopy
coupling was also achieved using a small molecule, 4PB
(Calbiochem, San Diego, CA, USA), which was previously
published to act as a gap-junction agonist, as well as shown to Immunofluorescence microscopy of vimentin (anti-chicken
increase Cx43 expression in cardiomyocytes (38). We therefore PA1-16759, 1:3,000; Thermo Fisher Scientific), a-SMA (anti-
added 1 mM 4PB to myofibroblasts alone or both cardiomyocytes mouse MA511547, 1:500; Thermo Fisher Scientific), Cx43
and myofibroblasts for 48 h at 37°C. 1-Heptanol was used to (anti-rabbit C6219, 1:1000; MilliporeSigma), and DAPI (33342,
completely uncouple gap junctions (39). Hypoxia leads to in- 1:1000; Thermo Fisher Scientific) were used to assess the inter-
ternalization of Cx43 (40). Hypoxia was induced by incubation nalization of Cx43 after hypoxia and the effect of latrunculin-B
at 37°C, 1% CO2, and 8% O2 overnight (;15 h). Single cultures on the heterocellular contact.
were seeded at least 24 h before hypoxia, whereas in cocultures,
myofibroblasts were seeded into 1–2-d-old cardiomyocytes ;4 h
before hypoxia treatment. For the dynamism experiments with Protein extraction and Western blots
hypoxia, cells were first treated with hypoxia, after which the
closed dish was taken out of the incubator and rapidly placed in Western blots for the gap-junctional protein, Cx43, were carried
the small, self-contained chamber that is necessary for recording. out as previously described (45); b-actin was used as a loading
We used each dish for a maximum of 1 h and did not find any control. In brief, cells were plated in 35-mm dishes (0.5 million

CARDIOMYOCYTE-MYOFIBROBLAST CX43-DEPENDENT MOVEMENT 3


g by Saab Medical Library, American Univ of Beruit (UAB) Levant U (193.188.128.21) on June 30, 2019. The FASEB Journal Vol. ${article.issue.getVolume()}, No. ${article.issue.get
Figure 1. SICM and the analysis
of contact movement. A) Sche-
matic of the SICM setup (left),
cartoons representing the dif-
ferent cell configurations used
here (top right), and the
analysis using CellTrack (44)
(bottom right). CellTrack (44)
selects a set of random points
(yellow squares) in the contact
area that is selected (circled)
and follows these throughout
the scans to calculate the move-
ment of the cell contact. B)
Representative images of a
CM–MFB contact, first scanned
with SICM (left) and subse-
quently with the laser confocal
(middle); an overlay showing
the junction (SICM scan) and
GFP-Cx43 in the junction
(green) is shown on the right.
C ) Representative set of scans
of the looped scans at represen-
tative time points. Each scan
takes 4–6 min, generating be-
tween 10 and 20 images, al-
though some may not be used
as debris on the cell may in-
terfere with the SICM scan. t,
time; z, cell height. D) Repre-
sentative image of CM–CM and
CM–MFB Cx43 distribution at the
cell–cell contacts. Red, b-catenin;
green, Cx43; pink, vimentin; blue,
DAPI. Cx43 was expressed at
the CM–MFB interface. The white
arrows indicate the presence of
Cx43 at the cell–cell contacts.
Scale bars, 10 mm.

cells per well) and cultured for 72 h with or without treatment. incubated on a shaker for 15 min on ice, and centrifuged for 15
The cells were subsequently washed with cold PBS and the min at 4°C, 14,000 g. Protein concentrations were determined
protein was extracted using lysis buffer containing protein in- using a BCA Protein Assay according to the manufacturer’s
hibitors. Collected lysates were sonicated for 1 min at 0°C, protocol (Pierce, Rockford, IL, USA). For Western blotting, each

4 Vol. 33 September 2019 The FASEB Journal x www.fasebj.org SCHULTZ ET AL.


g by Saab Medical Library, American Univ of Beruit (UAB) Levant U (193.188.128.21) on June 30, 2019. The FASEB Journal Vol. ${article.issue.getVolume()}, No. ${article.issue.get
protein sample was fractioned in denaturating conditions by RESULTS
SDS–PAGE electrophoresis. A Trans-Blot Turbo System (Bio-Rad,
Hercules, CA, USA) was used to transfer the proteins to the Dynamic measurement of cell–cell contacts
membrane (Bio-Rad). Membranes were blocked for 1 h in 5% milk
(MilliporeSigma) and then incubated overnight at 4°C in primary
We used neonatal rat cardiomyocytes and myofibroblasts
antibody (Cx43, C6219, anti-rabbit 1:1,000, MilliporeSigma;
b-actin, A3854, anti-mouse 1:50,000; MilliporeSigma) diluted in to establish a baseline (control) for the dynamism. After an
5% milk. The next day membranes were washed 3 times in area was identified to contain a cluster of both myofibro-
Tris-buffered saline with Tween-20 (TBST; 1 M Tris pH 8.0, NaCl blasts (identified by Vibrant DiI or WGA-488) and car-
diluted in distilled water) and incubated with horseradish per- diomyocytes (in the heterocellular cultures), or clusters of
oxidase (HRP)-conjugated secondary antibodies (7074, donkey either cell type (in homocellular cultures), a large scan was
anti-rabbit 1:1000, Cell Signaling Technology, Danvers, MA, obtained using SICM, followed by recording a set of scans
USA; 7076, donkey anti-mouse 1:1000; Cell Signaling Technology)
diluted in TBST for 1 h at room temperature. Blots were visualized
for 60 min. We then followed the contact over time and
using Luminata Forte Western HRP substrate (MilliporeSigma). analyzed the dynamism (see Materials and Methods). The
ImageJ software (NIH) was used to quantify bands that were presence of gap junctions at the cellular interfaces was
normalized to b-actin. confirmed using cardiomyocytes expressing Cx43 with
GFP fused to its C terminus (Cx43–GFP; Fig. 1). First, a
topographical image was obtained and subsequently, us-
Parachute assays ing confocal microscopy, the same area was imaged and
the Cx43–GFP was visualized. Each scan took ;5 min,
Myofibroblasts were plated on 35-mm dishes and, upon con- representative SICM and confocal images of Cx43–GFP at
fluency, treated with the indicated treatments. For 4PB experi-
ments, myofibroblasts were treated for 48 h to increase Cx43 the junction of a cardiomyocyte and myofibroblast
expression. To achieve Cx43 KD, myofibroblasts were either (CM–MFB) are shown in Fig. 1B. An example of the dis-
transfected with Cx43-specific siRNA or transduced with lentiviral tribution of Cx43 at the cardiomyocyte–cardiomyocyte
vector particles expressing a Cx43-specific shRNA to silence Cx43 (CM–CM) and CM–MFB contacts is presented in Fig. 1D.
and eGFP to detect the transfected cells, prior to the dye staining.
Cells were incubated with 5 mM Cell Tracker Orange/FarRed
(Thermo Fisher Scientific) and 1 mM Calcein/Calcein Orange Cx43-mediated coupling has implications for
(Thermo Fisher Scientific) for 30 min at 37°C, trypsinized and cell–cell border zone dynamism
plated on cardiomyocytes in a 1:2 ratio to form heterocellular
contacts. Cells were cocultured for 24 h. Live-cell imaging was We evaluated the dynamism of neonatal rat ventricular
performed on a Zeiss Laser Scanning Microscope 780 Confocal. CM–CM, CM–MFB, and myofibroblast–myofibroblast
Only cells that were calcein-positive and that were directly ad-
jacent to dually labeled fibroblasts (transferring cells) were (MFB–MFB) contacts (Fig. 2). The CM–CM pairs, known
counted. to be coupled by the electrical gap-junctional protein Cx43,
displayed minimal movement [0.04 6 0.005 mm/min, n =
14 scan sets (data were obtained from $2 dishes per iso-
Cell boundary measurement lation from n $ 3 isolations, unless otherwise indicated)],
whereas the border zone, modeled by the coculture of
Quantification of images was performed using a custom-made cardiomyocytes and myofibroblasts, showed a significant
computer vision program to identify cell–cell contact (courtesy of increase in dynamism (0.14 6 0.033 mm/min, n = 11 scan
V.B.). In brief, the cell boundary was selected based on the a-SMA
sets, P # 0.001). Interestingly, MFB–MFB contacts dis-
protein staining and manually calibrated. The threshold and di-
lation cycle measurements were set for the interface area to pick played a considerable level of dynamism in culture (0.44 6
up only Cx43 expressed at the cell boundary, and these were kept 0.22 mm/min, n = 7 scan sets, P # 0.05), compared to
constant throughout all image analyses. Cx43 labeling was heterocellular cell pairs.
identified and calculated as the intensity in Cx43 area (arbitrary
units/pixel2). Data are presented as means 6 SEM.
Membrane Cx43-deficient cells show
limited dynamism
Statistical analysis
The involvement of Cx43 in the CM–MFB and MFB–MFB
A statistical analysis was carried out in Prism 7 (GraphPad, La dynamism was elucidated by infecting myofibroblasts
Jolla, CA, USA) for all the scanning data and Western blots. All (or cardiomyocytes in CM–CM control experiments)
data are represented as means 6 SEM. All data were tested for
normality before a Kruskal–Wallis test with post hoc tests (scans,
with lentiviral vector particles encoding GFP and a Cx43-
Western blotting) or Student’s t test (parachute assays) was carried specific shRNA (Cx43–GFP shRNA) or by using siRNA
out. For all scanning data, n represents the number of 1 h scan sets against Cx43 (Fig. 3). Green fluorescence was used to
that were used. More image sets were carried out but may not have identify myofibroblasts in which Cx43 was successfully
been used for various reasons, including noise in the scan due to silenced. The myofibroblast membrane was also labeled
debris on the cell, which interferes with the scan of the membrane. with a lipophilic membrane dye (Vybrant DiI, Thermo
All data were carried out with at least 1 dish of cells; dishes were Fisher Scientific; see Supplemental Fig. S1). Cx43 KD sig-
changed after 1–2 scan sets, especially for the hypoxia work, as the
scans were carried out at normal oxygen tension, which meant that nificantly slowed the cell–cell dynamism of contacts in-
changes induced by hypoxia may have reverted back after 1–2 h. volving myofibroblasts: CM–MFB (0.017 6 0.013 mm/
Most experiments were carried out with cells from at least 2 sep- min, n = 6 scan sets, n = 2 isolations, P # 0.01) and
arate isolations (10–15 pups/isolation). MFB–MFB (0.027 6 0.007 mm/min, n = 7 scan sets, n = 2

CARDIOMYOCYTE-MYOFIBROBLAST CX43-DEPENDENT MOVEMENT 5


g by Saab Medical Library, American Univ of Beruit (UAB) Levant U (193.188.128.21) on June 30, 2019. The FASEB Journal Vol. ${article.issue.getVolume()}, No. ${article.issue.get
Figure 2. Dynamism in control CM–CM, CM–MFB, and MFB–MFB contacts. A) Representative set of scans for each
configuration. Cartoons indicate the configuration, as shown in Fig. 1. The red line is included for the contact between
MFB–MFB for clarity. t, time; z, cell height. B) Movement speed of the contacts. All data are represented as means 6 SEM. *P #
0.05, ***P # 0.001.

isolations, P # 0.01) but did not significantly affect car- the myofibroblast membrane, and not their function, is
diomyocytes alone [0.035 6 0.012, n = 5 scan sets (from a required to activate the dynamic coupling (Supplemental
single isolation)] (Fig. 3A, B). By contrast, 50 mM heptanol Fig. S2).
(a gap-junctional uncoupler) did not slow the dynamism To confirm that the gap junctions between car-
in CM–MFB (0.047 6 0.010, n = 7 scan sets, n = 2 isolations) diomyocytes and myofibroblasts were functional and to
and MFB–MFB (0.059 6 0.016, n = 6 scan sets, n = 2 iso- investigate the extent of direct physical coupling, para-
lations), suggesting that the presence of gap junctions on chute assays were carried out (Fig. 3C, D). Control acceptor

6 Vol. 33 September 2019 The FASEB Journal x www.fasebj.org SCHULTZ ET AL.


g by Saab Medical Library, American Univ of Beruit (UAB) Levant U (193.188.128.21) on June 30, 2019. The FASEB Journal Vol. ${article.issue.getVolume()}, No. ${article.issue.get
Figure 3. Cx43 KD leads to
decreased dynamism and re-
duced dye transfer. A) Repre-
sentative set of scans for each
configuration. Illustrations in-
dicate the configuration, as
shown in Fig. 1. t, time; z, cell
height. B) Movement speed of
the contacts after Cx43 KD
using siRNA in only the myofi-
broblasts (in CM–MFB cultures)
or in either cell type in the
monocultures. C–E ) Cx43 KD
using Cx43-specific siRNA or
Cx43–GFP shRNA in only the
myofibroblasts leads to de-
creased dye transfer and there-
fore a reduced number of
functional gap junctions. C )
Analysis of the parachute assay.
Cx43 siRNA, n = 52 images (for
all parachute assays: the total
number of images from 2 dishes
per isolation for n = 3 isolations
are indicated); control, n = 61
images. Cx43–GFP shRNA, n =
29 images; control, n = 29
images. D) Representative im-
ages of control and Cx43 siRNA.
Calcein (green) is used as a
cell-permeable dye, whereas
CellTracker Orange (red) is a
cell-impermeable dye. E ) Rep-
resentative images of control
and Cx43–GFP shRNA. GFP
(green) is used to identify
myofibroblasts with Cx43 KD (only
shown in the shRNA-treated im-
ages). Calcein Orange (orange) is
used as a cell-permeable dye,
whereas CellTracker FarRed
(magenta) is a cell-impermeable
dye. Scale bars, 100 mm. All data
are represented as means 6 SEM.
*P # 0.05, **P # 0.01, ***P #
0.001.

cells (cardiomyocytes) were plated and control or myofibroblasts had been dually labeled with permeable
Cx43-deficient myofibroblasts [Cx43 siRNA (Fig. 3D, bot- (Calcein or Calcein Orange) and impermeable (CellTracker
tom left) or Cx43–GFP shRNA (Fig. 3D, bottom right)] were Orange or CellTracker FarRed) dyes. Myofibroblasts with
added to cultures of cardiomyocytes. Prior to addition, reduced Cx43 expression due to treatment with either Cx43

CARDIOMYOCYTE-MYOFIBROBLAST CX43-DEPENDENT MOVEMENT 7


g by Saab Medical Library, American Univ of Beruit (UAB) Levant U (193.188.128.21) on June 30, 2019. The FASEB Journal Vol. ${article.issue.getVolume()}, No. ${article.issue.get
siRNA [50.50 6 6.457%, n = 52 images (for all parachute with the small molecule (0.063 6 0.012, n = 29, P # 0.05), but
assays: the total number of images from 2 dishes per iso- not when only the myofibroblasts were incubated with 4PB
lation for n = 3 isolations are indicated); control, 100 6 (0.183 6 0.086, n = 55). Using an in vitro assay of direct
6.236%, n = 61 images; P # 0.001] or Cx43–GFP shRNA cell–cell communication in the parachute assay, there is a
(55.63 6 2.662%, n = 29 images; control, 100 6 7.333%, n = 29 significant increase in dye transfer in both cases; 4PB
images; P # 0.001) showed significantly attenuated calcein CM–MFB: 159.3 6 10.64%, n = 29 vs. control (100 6 8.479%,
transfer compared to the respective control conditions n = 33) and myofibroblasts only treated with 4PB: 159.3 6
(normalized to 100%). 9.961%, n = 55 vs. control (100 6 7.425%, n = 53) (Fig. 4A, B).
To confirm KD of Cx43 and to investigate whether 4PB
has a similar effect on myofibroblasts as was previously
Increased cellular coupling induced by 4PB published in cardiomyocytes (38), Western blotting was
carried out (Fig. 4D, E). 4PB treatment did not lead to a
A small molecule (4PB) has previously been shown to act as significant increase in Cx43 levels in cardiomyocytes or
an epigenetic modulator that increases cellular functional myofibroblasts, in contrast to previous findings in car-
coupling between cardiomyocytes (38). We used this mol- diomyocytes (38). However, this may be caused by dif-
ecule as a method to strengthen Cx43 coupling between ferences in incubation time or technical repeats (n = 3–4)
cardiomyocytes and myofibroblasts (Fig. 4). 4PB induced a and a potentially lower baseline level of Cx43 expression
significant reduction in dynamism in the CM–MFB con- in myofibroblasts compared to cardiomyocytes. Fol-
figuration (Fig. 4C), when both cell types were incubated lowing Cx43 KD, Cx43 protein levels were reduced in

Figure 4. Increased cellular


coupling induced by 4PB. A)
Representative images of con-
trol cocultures and cocultures
with 4PB-treated myofibroblasts.
Calcein (green) is used as
a cell-permeable dye, whereas
CellTracker Orange (red) is a
cell-impermeable dye. Scale
bars, 100 mm. B) Analysis of the
parachute assay. 4PB treatment
of cocultured cardiomyocytes
and myofibroblasts (n = 29
images, n = 3 isolations) or
myofibroblasts alone (n = 55
images, n = 3 isolations) leads
to increased calcein transfer. C )
Treatment of both cardiomyo-
cytes and myofibroblasts, but not
myofibroblasts alone, leads to
reduced movement speed in
the contact. D, E) Western blot
analysis of Cx43 (n = 3–4 in-
dependent isolations). Quantifi-
cation (D) and representative
blots (E) are shown. AU, arbi-
trary units. All data are repre-
sented as means 6 SEM. *P #
0.05, ***P # 0.001.

8 Vol. 33 September 2019 The FASEB Journal x www.fasebj.org SCHULTZ ET AL.


g by Saab Medical Library, American Univ of Beruit (UAB) Levant U (193.188.128.21) on June 30, 2019. The FASEB Journal Vol. ${article.issue.getVolume()}, No. ${article.issue.get
cardiomyocytes to 69% of control (P = 0.057), and although cell-permeable dye Calcein and cell-impermeable dye Cell-
not significant, a trend toward reduction was found in Tracker Orange and treated with either the aCT1 or reverse
myofibroblasts; Cx43 expression was reduced to 41% of (control) peptide. The myofibroblasts were then parachuted
control values (n = 3–4, P = 0.094; Fig. 4D, E). As Cx43 onto cardiomyocytes, which were also treated with either the
expression is generally lower in myofibroblasts compared aCT1 or reverse peptide (Fig. 5G; n = 4 isolations/treatment).
to cardiomyocytes (35, 46), this reduction was found to be Although not significant, a small decrease in gap junction
sufficient to induce the observed changes in membrane intercellular communication, after treatment with the aCT1
motility and calcein transfer. peptide, was observed (P = 0.12) with respect to control.

Hypoxia-induced internalization of Cx43 Actin filament-based modification of Cx43


induced a decrease in dynamism
Myofibroblasts are known to promote conduction slowing
The presence of Cx43 on the membranes of isolated car- and ectopic activity in cardiac tissue (25, 47). As previously
diomyocytes and myofibroblasts was assessed by cultur- shown (41), these arrhythmogenic properties can be sup-
ing the cells in hypoxic conditions (8% O2) for 12–16 h pressed by disruption of a-SMA stress fibers using phar-
before measuring cell–cell dynamism (Fig. 5B). Hypoxia is macological manipulation. Moreover, trafficking of Cx43
known to internalize the connexons in the plasmalemma of to the membrane is mediated by a-actin filaments. To in-
cardiomyocytes (40) and myofibroblasts (Fig. 5B, E and vestigate the role of a-SMA in cell membrane mobility
Supplemental Fig. S3C). As postulated, for both CM–MFB and to prevent intercellular gap-junctional communication
[0.067 6 0.021, n = 7 scan sets (3 dishes each from 2 isola- and functional Cx43 connexon expression on the sarco-
tions), P # 0.05] and MFB–MFB (0.100 6 0.025, n = 13 scan lemma, neonatal CM–MFB cocultures were treated with
sets, P # 0.05) models, limited dynamism was measured latrunculin-B, an inhibitor of actin polymerization. After
when cells were cultured in hypoxic conditions compared incubation for 24 h, the myofibroblast dynamism was sig-
to normoxic (control) conditions. This indicates that the nificantly decreased [0.03 6 0.006 mm/min, n = 9 scan sets
presence of Cx43, and thus the capacity to establish cell–cell (2 and 4 dishes from 2 isolations), P # 0.05; Fig. 6A, C]. Cell
electrical coupling, is necessary for cell–cell border zone membrane mobility was then examined in myofibroblasts
movement. In terms of functional coupling, chronic hyp- isolated from a rat model of myocardial infarction (16 wk
oxia also caused a reduction in dye transfer from neonatal postinfarction) (32). a-SMA ablation resulted in lower cell
myofibroblasts to neonatal cardiomyocytes (Supplemental dynamism (0.03 6 0.004 mm/min, n = 13 scan sets) com-
Fig. S3A, B). pared to untreated myofibroblasts from the same model
(0.12 6 0.041 mm/min, P # 0.01; Fig. 6E, G). We assessed the
functionality of CM–MFB contacts in both models using a
Inducing recruitment of Cx43 to the cell
parachute assay and observed a significant reduction in
membrane using the mimetic aCT1 peptide
calcein transfer following treatment with latrunculin-B in
cocultures of neonatal cardiomyocytes and myofibroblasts
A second approach was used to examine the effects of
(latrunculin-B, 20.18 6 5.113%, n = 38 images, n = 3; control,
increased levels of Cx43-mediated contact. The mimetic
Cx43 peptide aCT1 has previously been shown to pro- 100 6 4.744%, n = 35 images, n = 3; P # 0.001; Fig. 6B, D) and
among adult rats with myocardial infarction (latrunculin-B,
mote the extent of Cx43 gap junction-mediated contacts
25.8 6 3.8%, n = 14 images, n = 3; control, 100 6 10.36%, n =
between cells (37), as well as reducing inducible arrhyth-
14 images, n = 3; P # 0.001) (Fig. 6F, H). Similar to the
mias at the CM–MFB zone following ventricular cry-
decrease in calcein transfer seen after latrunculin-B treat-
oinjury (19). We therefore investigated the effect of the
ment of myofibroblast cultures derived from adult rats with
peptide in our cellular heterocellular model (Fig. 5). In both
myocardial infarction, control (sham) rat myofibroblast
cardiomyocyte and myofibroblast monocultures, as well
cultures also showed a reduction (42%, P , 0.001) in dye
as in cocultures of these cells, the aCT1 peptide induced
transfer after latrunculin-B treatment (Supplemental Fig.
the occurrence of Cx43 at the contact regions between cells.
S4C, D); this was accompanied by a decrease in cell dyna-
The increased presence of Cx43-mediated contacts in-
mism (Supplemental Fig. S4A, B). This indicates that
creased CM–MFB dynamism [0.415 6 0.090 mm/min, n =
5 scan sets (1–2 dishes from 3 isolations)], indicating a myofibroblasts derived from both adult rats with myo-
cardial infarction and sham-treated adult rats are sensitive
novel “mechanical” role for an “electrical” intercellular
to the effects of latrunculin-B. Significant effects of both
connection (Fig. 5C, F). By contrast, hypoxic conditions, in
condition [sham vs. myocardial infarction (P = 0.0006)] and
the presence of the aCT1 peptide [0.057 6 0.015 mm/min,
treatment [control vs. latrunculin-B (P = 0.0364)] on contact
n = 6 scan sets (3 dishes each from 2 isolations)], or hypoxia
dynamism was found (2-way ANOVA), but the interaction
after a 4-h preincubation with the aCT1 peptide [0.050 6
between treatment and condition was not significant.
0.018 mm/min, n = 3 (3 dishes from a single isolation)],
significantly reduced dynamism (Fig. 5) despite Cx43 lo-
calization at heterocellular junctions (data not shown). Changes in the level of Cx43 expression in the
Therefore, we investigated whether enhanced Cx43- different treatment groups
mediated coupling had a functional role in intercellular
communication between cardiomyocytes and myofibro- In order to further validate that the changes seen were due
blasts (Fig. 5G). Myofibroblasts were dually labeled with the to the functional effects of Cx43, protein levels were

CARDIOMYOCYTE-MYOFIBROBLAST CX43-DEPENDENT MOVEMENT 9


g by Saab Medical Library, American Univ of Beruit (UAB) Levant U (193.188.128.21) on June 30, 2019. The FASEB Journal Vol. ${article.issue.getVolume()}, No. ${article.issue.get
Figure 5. Removing Cx43 by hypoxia and forcing Cx43 to the membrane by aCT1 peptide. A–D) Representative scans of control
and treatments. z, cell height. E ) Hypoxia reduces the dynamism between cells. F ) aCT1 treatment leads to increased dynamism
in control, but not in hypoxic, conditions. G) Treatment with the aCT1, but not the reverse, peptide leads to decreased dye
transfer, showing that, although the plaque size increases, the number of functional gap junctions decreases. All data are
represented as means 6 SEM. *P # 0.05, ***P # 0.001. For representative Western blots and quantification of Cx43 protein
expression in cardiomyocytes and myofibroblasts treated with hypoxia or aCT1, see Fig. 7A, B.

10 Vol. 33 September 2019 The FASEB Journal x www.fasebj.org SCHULTZ ET AL.


g by Saab Medical Library, American Univ of Beruit (UAB) Levant U (193.188.128.21) on June 30, 2019. The FASEB Journal Vol. ${article.issue.getVolume()}, No. ${article.issue.get
Figure 6. Treatment with latrunculin-B abolishes contact dynamism and dye transfer. A, B) Representative SICM scans (A) and images of
parachute assays (B) of control CM–MFB cultures and CM–MFB cultures treated with latrunculin-B from neonatal rats. z, cell height. C,
D) Treatment with latrunculin-B leads to reduced dynamism (C) and a reduction in the number of functional contacts (D). B, D,
Control, n = 35 images, n = 3 isolations; Latrunculin-B, n = 39 images, n = 3 isolations. For representative Western blots and quantification
of Cx43 protein expression in cardiomyocytes and myofibroblasts treated with latrunculin-B, see Fig. 7A, B. E, F) Representative SICM
scans (E) and images of parachute assays (F) of myofibroblasts cultures derived from adult rats with myocardial infarction that were
treated with latrunculin-B or vehicle control (MFB–MFB). For experiments carried out using myofibroblasts from sham-treated adult
rats, see Supplemental Fig. S4. G, H) Treatment with latrunculin-B leads to reduced dynamism (G) and a reduction in the number of
functional contacts (H). F, H, Control, n = 24 images, n = 3 isolations; Latrunculin-B, n = 25 images, n = 3 isolations. CTO, CellTracker
Orange; LatB, latrunculin-B. All data are represented as means 6 SEM. Scale bars, 100 mm. *P # 0.05, **P # 0.01, ***P # 0.001.

CARDIOMYOCYTE-MYOFIBROBLAST CX43-DEPENDENT MOVEMENT 11


g by Saab Medical Library, American Univ of Beruit (UAB) Levant U (193.188.128.21) on June 30, 2019. The FASEB Journal Vol. ${article.issue.getVolume()}, No. ${article.issue.get
assessed in separate cultures of neonatal cardiomyocytes DISCUSSION
and fibroblasts using Western blot. Hypoxic conditions
reduce the levels of Cx43 in both cell types. A marked drop Pathologic events, such as myocardial infarction, trigger
in Cx43 expression is also observed in the myofibroblast complex changes and dynamic remodeling of the cardiac
cultures treated with latrunculin-B. By contrast, aCT1 tissue, which occurs over time. Through coordinated reg-
treatment notably increased the level of the Cx43 expres- ulation of the gap-junctional proteins and the extracellular
sion in myofibroblast samples (Fig. 7A, B). matrix in the affected region, the irreversibly damaged
Immunofluorescence staining was performed to visu- myocardium becomes electrically quarantined from the
alize the contacts that had formed. Heterocellular connec- rest of the heart to restrict the spread of adverse effects; the
tions were further quantified using a custom-made cell border zone area between the infarct and the healthy tissue
boundary measurement tool (V.B. laboratory). Quantify- undergoes highly dynamic changes in cell–cell commu-
ing the Cx43 intensity in the Cx43 area, we observed that nication. It has been reported that myofibroblasts coupled
the disruption of a-SMA reduced the functionality of to cardiomyocytes show an increase in Cx43 expression
CM–MFB Cx43-based coupling compared to the untreated and an associated increase in gap junction formation (21).
sample (n = 15 images, n = 3 isolations, P # 0.01; Fig. 7A, B), Cx43-based contact between myofibroblasts and car-
which is consistent with the parachute assay data of cul- diomyocytes is thought to contribute to ectopic activity and
tures treated with latrunculin-B. arrhythmias (25, 47). In the present study, we show a novel

Figure 7. Quantification of Cx43


expression in control, hypoxia-,
latrunculin-B-, and aCT1-treated
samples. A) Cx43 and loading
control [glyceraldehyde 3-phos-
phate dehydrogenase (GAPDH)]
intensity bands in neonatal
cardiomyocyte and myofibroblast
cultures (n 5 2–4 independent
isolations). B) Quantification of
Western blots shown in A. All
data are represented as means 6
SEM; n = 2–4 independent isola-
tions. C ) Latrunculin-B treat-
ment significantly decreases the
Cx43 intensity in the Cx43
clusters (n = 15 images, n = 3
isolations). All data are repre-
sented as means 6 SEM. **P #
0.01. D) Representative images
of immunostaining of CM–MFB
contacts. Red, a-SMA; green,
Cx43; pink, vimentin; blue, DAPI.
Cx43 was expressed at the
CM–MFB interface. Scale bars,
10 mm. AU, arbitrary units;
latrunculin-B, LatB.

12 Vol. 33 September 2019 The FASEB Journal x www.fasebj.org SCHULTZ ET AL.


g by Saab Medical Library, American Univ of Beruit (UAB) Levant U (193.188.128.21) on June 30, 2019. The FASEB Journal Vol. ${article.issue.getVolume()}, No. ${article.issue.get
Cx43-dependent aspect of myofibroblast behavior that may recognized action as a chemical chaperone, which can re-
contribute to heart failure pathology. Specifically, myofi- duce endoplasmic reticulum stress and can attenuate the
broblasts demonstrate highly motile contacts with each unfolded protein response (50, 51). Better folding and im-
other and with cardiomyocytes. This unique dynamism is proved trafficking to the membrane can give rise to better
characterized by the rapid formation of new areas of contact and more stable (low dynamism) electrical coupling even if
and similarly efficient remodeling of existing interactions. Cx43 transcription is not significantly changed, which is
By contrast, cardiomyocytes in culture form stable gap probably the case here.
junctions with each other and have very low movement in This suggests that there may be an optimal level of
zones of mutual Cx43-based interactions—consistent with Cx43-mediated electrical coupling that allows for high dy-
the idea that myocardial cell-to-cell coupling is important namism of CM–MFB contacts. When Cx43 levels are too low
for the propagation of electrical impulse (48). in the contact area, the edge motility of myofibroblasts and
This dynamic ability to remodel cell–cell contacts appears heterocellular contact dynamics are reduced. When the
to be necessary for the supportive roles of myofibroblasts. electrical coupling is too high in the contact area, this becomes
An interesting question arises as to what the relationship too restrictive and dynamism is also reduced. Myofibroblasts
between these physical contact dynamics and the drivers of are normally only lightly coupled to each other and to car-
electrical coupling, Cx43 gap junctions, is. In this study, us- diomyocytes, with contacts breaking and reforming contin-
ing a variety of methods to modulate assembly of Cx43 and uously; they are highly mobile and show a lot of plasticity.
actin, we aimed to address this question. Our results indicate For instance, in vitro studies have shown that they are capa-
that this relationship (contact dynamism and electrical ble of bridging gaps among cardiomyocytes of up to 300 mm
coupling) is not simple and monotonic. The levels of (15, 21). Increasing the number of Cx43 connexons on the
gap-junctional interaction require tight control to ensure myofibroblast membrane that are effectively coupled to their
that, while sufficient levels of intercellular coupling are in counterparts in the cardiomyocyte membrane increases the
place, gap junction-mediated adhesions are not so strong as strength of the CM–MFB contacts, and as the pair starts be-
to restrict the dynamism of heterocellular contacts. Follow- having as one, the motility of the myofibroblast is reduced.
ing myocardial infarction, myofibroblasts showed an in- Treatment with the aCT1 peptide has been shown to lead
crease in Cx43 expression and associated level of gap to an increase in Cx43 plaque size and a decrease in Cx43
junction formation (21). hemichannels (37, 52). Although treatment with the aCT1
In our experiments, CM–MFB cultures exhibited much peptide led to an increase in dynamism, this was associated
higher membrane contact dynamics than cardiomyocyte with a small decrease in dye transfer. This can be explained
monocultures. This difference in the vigor of cell–cell by the fact that, after treatment of the cells with the aCT1
membrane contact dynamics was associated with differ- peptide, the phosphorylation status of Cx43 is likely to
ences in Cx43 expression and the stability of intercellular have changed, as phosphorylation is one of the main ways
Cx43 connections. We show that, although myofibroblasts in which Cx43 channels are regulated through post-
had lower Cx43 expression (35) than cardiomyocytes, Cx43 translational modification (53–55). We have previously re-
functionally couples cardiomyocytes and myofibroblasts, ported that aCT1 increased Cx43 phosphorylation at S368 at
as indicated by intercellular calcein transfer. It appears that the interface between cardiomyocyte and myofibroblast in
Cx43 serves both as an affector and as a sensor of contact vivo (19), a post-translational modification known to be as-
dynamism between cells. Furthermore, we determined that sociated with decreased Cx43 channel activity (56). We have
decreasing the level of Cx43 through Cx43 silencing, by observed that aCT1 similarly increased S368 phosphoryla-
preventing Cx43 trafficking, or through Cx43 internaliza- tion in our in vitro CM–MFB model (57). The observed re-
tion reduced contact dynamism. duction in dye transfer between coupled cells is therefore
This is correlated with a reduction in dye transfer and, likely to be explained by this change in Cx43 phosphoryla-
thus, a reduction in gap-junctional coupling between tion status, despite an increase in Cx43 expression and plaque
myofibroblasts and cardiomyocytes. By contrast, increas- size. In addition, with recruitment of Cx43 to the membrane,
ing membrane Cx43 levels did not necessarily lead to an myofibroblasts treated with the aCT1-peptide showed in-
increase in coupling and dynamism. We found that 4PB creased lamellipodia formation, which may indicate their
increased coupling between cardiomyocytes and myofi- search for stable connections with neighboring cells.
broblasts without changing Cx43 protein levels (Fig. 4); It may be hypothesized that other modifications, such
however, interestingly, it decreased dynamism. as ubiquitination (which would decrease the amount of
In human glioblastoma cells, it was shown that levels of protein at the junctions) are reduced or changed (for ex-
unphosphorylated Cx43 were increased after treatment ample, acetylation or methylation) after treatment with
with 4PB, whereas the phosphorylated form remained sta- our interventions; however, it is beyond the scope of this
ble (49). We assume that the stabilization at the membrane study to analyze these modifications further.
that we have previously shown (38) occurs through a It has been shown that Cx43 trafficking to the sarco-
combined increase of both forms. Notably, 4PB affects Cx43 lemma of cardiomyocytes is dependent on both the mi-
epigenetically; therefore, there is not necessarily a change in crotubular network (58) and actin cytoskeleton (59).
expression. 4PB is known to exert multiple effects. 4PB is Previously, the role of a-SMA stress fibers in myofibro-
an epigenetic modulator that increases cellular functional blasts has been investigated, and it has been shown that
coupling between 2 cardiomyocytes (unpublished data), pharmacological ablation of these cytoskeletal elements
2 myofibroblasts or between a cardiomyocyte and myofi- eliminates the arrhythmogenic effects of myofibroblasts
broblast. Among the nonepigenetic effects of 4PB is its on cardiomyocytes (12, 41, 59). We therefore expanded on

CARDIOMYOCYTE-MYOFIBROBLAST CX43-DEPENDENT MOVEMENT 13


g by Saab Medical Library, American Univ of Beruit (UAB) Levant U (193.188.128.21) on June 30, 2019. The FASEB Journal Vol. ${article.issue.getVolume()}, No. ${article.issue.get
this research by studying the effect of pharmacological How the Cx43-dependent myofibroblast dynamism
ablation of the actin cytoskeleton in cocultures of car- characterized in our study affects the formation of proar-
diomyocytes and myofibroblasts to investigate the impact rhythmic substrates at the heterocellular coupling area
on heterocellular contact dynamism. Disruption of the should be investigated further. The irregularity of the in-
actin cytoskeleton significantly reduced CM–MFB contact terface between surviving cardiac muscle and the scar tissue
movement in cocultures of neonatal rat cardiomyocytes of myocardial infarction is a well-characterized determinant
with either neonatal rat myofibroblasts or myofibroblasts and originator of re-entrant arrhythmias (60, 61). Modeling
derived from adult rats with myocardial infarction. Sub- studies have shown that microstructural variations in car-
sequently, we investigated the diminishing heterocellular diac tissue facilitate the formation of isolated sites of wave-
contact functionally. Using a custom-made cell–cell mea- front breakthrough that may enable abnormal electrical
surement software (V.B. laboratory), we found a partial activity; these small regions of heterogeneous diseased tissue
drop in Cx43 intensity in the Cx43 area after a-SMA stress can develop more widespread re-entrant activity (62, 63).
fiber removal, which could be related to the actin-dependent The relationship between Cx43 expression and level of
Cx43 pool that has previously been described (59). These heterocellular contact dynamism that we report here likely
results indicate that Cx43 not only affects contact dyna- has implications for the disposition of mechanical forces
mism, but also that the opposite is true—Cx43 coupling imparted by myofibroblasts on cardiomyocytes. How
senses and reacts to actin dynamics. Cx43-based mechano-interactions at the microscale shape

Figure 8. Schematic of our current view on the role of Cx43 in CM–MFB dynamism. Max., maximum; min., minimum.

14 Vol. 33 September 2019 The FASEB Journal x www.fasebj.org SCHULTZ ET AL.


g by Saab Medical Library, American Univ of Beruit (UAB) Levant U (193.188.128.21) on June 30, 2019. The FASEB Journal Vol. ${article.issue.getVolume()}, No. ${article.issue.get
the macrostructure of the extended heterocellular interface 3. Alvarez-Maubecin, V., Garcia-Hernandez, F., Williams, J. T., and
thus may be of key importance to understanding the for- Van Bockstaele, E. J. (2000) Functional coupling between neurons
and glia. J. Neurosci. 20, 4091–4098
mation and pathogenesis of arrhythmic re-entrant circuits. 4. Haefliger, J. A., Nicod, P., and Meda, P. (2004) Contribution of
In summary, here we reveal the complex relationship connexins to the function of the vascular wall. Cardiovasc. Res. 62, 345–356
between Cx43-mediated heterocellular electrical coupling 5. Truskey, G. A. (2010) Endothelial cell vascular smooth muscle cell
co-culture assay for high throughput screening assays for discovery of
and physical contact dynamism (Fig. 8). We show that anti-angiogenesis agents and other therapeutic molecules. Int. J. High
Cx43 is not only involved in electrical coupling between Throughput Screen. 2010, 171–181
cardiomyocytes and myofibroblasts, but is also important 6. Hirschi, K. K., Rohovsky, S. A., and D’Amore, P. A. (1998) PDGF,
for the mechanical properties of the contacts. We provide TGF-b, and heterotypic cell-cell interactions mediate endothelial
cell-induced recruitment of 10T1/2 cells and their differentiation to a
new insights into Cx43-mediated coupling between car- smooth muscle fate. J. Cell Biol. 141, 805–814; erratum: 1287
diomyocytes and nanoscale evidence of the formation and 7. Rhee, S. (2009) Fibroblasts in three dimensional matrices: cell
dynamism of heterocellular contacts. Mechanistically, we migration and matrix remodeling. Exp. Mol. Med. 41, 858–865
demonstrate that Cx43 is necessary for both the dynamism 8. Pinto, A. R., Ilinykh, A., Ivey, M. J., Kuwabara, J. T., D’Antoni, M. L.,
Debuque, R., Chandran, A., Wang, L., Arora, K., Rosenthal, N. A., and
of structural membrane contacts and for direct physical Tallquist, M. D. (2016) Revisiting cardiac cellular composition. Circ.
coupling of myofibroblast and cardiomyocytes in an in Res. 118, 400–409
vitro infarct border zone model. 9. Gourdie, R. G., Dimmeler, S., and Kohl, P. (2016) Novel therapeutic
strategies targeting fibroblasts and fibrosis in heart disease. Nat. Rev.
Drug Discov. 15, 620–638
10. Kuwahara, F., Kai, H., Tokuda, K., Kai, M., Takeshita, A., Egashira, K.,
ACKNOWLEDGMENTS and Imaizumi, T. (2002) Transforming growth factor-b function
blocking prevents myocardial fibrosis and diastolic dysfunction in
The authors thank P. O’Gara (Imperial College London, pressure-overloaded rats. Circulation 106, 130–135
London, United Kingdom) for the assistance with the isolation of 11. Shinde, A. V., and Frangogiannis, N. G. (2014) Fibroblasts in
adult rat ventricular fibroblasts and M. Sikkel (Imperial College myocardial infarction: a role in inflammation and repair. J. Mol.
London, London) and C. Mansfield (Imperial College London, Cell. Cardiol. 70, 74–82
London) for generating rat models of myocardial infarction. This 12. Smyth, J. W., Hong, T.-T., Gao, D., Vogan, J. M., Jensen, B. C., Fong,
work was supported by British Heart Foundation (RG/17/13/ T. S., Simpson, P. C., Stainier, D. Y. R., Chi, N. C., and Shaw, R. M. (2010)
33173 to J.G.), Medical Research Council (MR/L006855/1 to Limited forward trafficking of connexin 43 reduces cell-cell coupling in
stressed human and mouse myocardium. J. Clin. Invest. 120, 266–279
J.G.), National Heart, Lung, and Blood Institute (NHLBI), U.S. 13. Foo, I. T., Naylor, I. L., Timmons, M. J., and Trejdosiewicz, L. K. (1992)
National Institutes of Health Grant-1R01HL141855-01 (to R.G., Intracellular actin as a marker for myofibroblasts in vitro. Lab. Invest.
J.G. and A.A.-L.), Heart Research UK, and by Italian Ministry of 67, 727–733
Education, Universities and Research (FFABR-MIUR-2017 to 14. Miragoli, M., Gaudesius, G., and Rohr, S. (2006) Electrotonic
M.M.). M.M. and J.G. jointly supervised this work. The authors modulation of cardiac impulse conduction by myofibroblasts. Circ.
declare no conflicts of interest. Res. 98, 801–810
15. Gaudesius, G., Miragoli, M., Thomas, S. P., and Rohr, S. (2003)
Coupling of cardiac electrical activity over extended distances by
fibroblasts of cardiac origin. Circ. Res. 93, 421–428
AUTHOR CONTRIBUTIONS 16. Biernacka, A., and Frangogiannis, N. G. (2011) Aging and cardiac
fibrosis. Aging Dis. 2, 158–173
M. Miragoli and J. Gorelik conceived the study; F. Schultz, 17. Rohr, S. (2009) Myofibroblasts in diseased hearts: new players in
P. Swiatlowska, A. Alvarez-Laviada, M. Miragoli, and J. Gorelik cardiac arrhythmias? Heart Rhythm 6, 848–856
designed experiments; F. Schultz, P. Swiatlowska, 18. McAnulty, R. J. (2007) Fibroblasts and myofibroblasts: their source,
function and role in disease. Int. J. Biochem. Cell Biol. 39, 666–671
A. Alvarez-Laviada, and M. Miragoli isolated, cultured, and 19. O’Quinn, M. P., Palatinus, J. A., Harris, B. S., Hewett, K. W., and
treated the cells; F. Schultz, P. Swiatlowska, and M. Miragoli Gourdie, R. G. (2011) A peptide mimetic of the connexin43 carboxyl
carried out scanning ion conductance microscopy (SICM) terminus reduces gap junction remodeling and induced arrhythmia
experiments; A. Alvarez-Laviada and J. L. Sanchez-Alonso following ventricular injury. Circ. Res. 108, 704–715
carried out SICM/confocal experiments; A. Alvarez-Laviada 20. Rubart, M., Tao, W., Lu, X.-L., Conway, S. J., Reuter, S. P., Lin, S.-F.,
and Soonpaa, M. H. (2018) Electrical coupling between ventricular
and E. Ongstad performed parachute assays; F. Schultz, myocytes and myofibroblasts in the infarcted mouse heart. Cardiovasc.
P. Swiatlowska, A. Alvarez-Laviada, Q. Song, and M. Miragoli Res. 114, 389–400
analysed the data; F. Schultz, P. Swiatlowska, and M. Miragoli 21. Camelliti, P., Devlin, G. P., Matthews, K. G., Kohl, P., and Green, C. R.
carried out Western blots; A. A. F. de Vries, D. A. Pijnappels, (2004) Spatially and temporally distinct expression of fibroblast
V. M. M. Braga, E. Entcheva, and R. G. Gourdie provided connexins after sheep ventricular infarction. Cardiovasc. Res. 62, 415–425
22. Van Veen, A. A., van Rijen, H. V. M., and Opthof, T. (2001) Cardiac
advice or supervision, assisted in analysis, provided reagents gap junction channels: modulation of expression and channel
and commented on the manuscript; and F. Schultz wrote properties. Cardiovasc. Res. 51, 217–229
the manuscript with input from P. Swiatlowska, A. Alvarez- 23. Miragoli, M., Moshkov, A., Novak, P., Shevchuk, A., Nikolaev, V. O.,
Laviada, J. L. Sanchez-Alonso, V. M. M. Braga, E. Entcheva, El-Hamamsy, I., Potter, C. M. F., Wright, P., Kadir, S. H. S. A., Lyon,
R. G. Gourdie, M. Miragoli, and J. Gorelik. A. R., Mitchell, J. A., Chester, A. H., Klenerman, D., Lab, M. J.,
Korchev, Y. E., Harding, S. E., and Gorelik, J. (2011) Scanning ion
conductance microscopy: a convergent high-resolution technology
for multi-parametric analysis of living cardiovascular cells. J. R. Soc.
REFERENCES Interface 8, 913–925
24. Miragoli, M., Salvarani, N., and Rohr, S. (2007) Myofibroblasts induce
1. Kohl, P., and Gourdie, R. G. (2014) Fibroblast-myocyte electrotonic ectopic activity in cardiac tissue. Circ. Res. 101, 755–758
coupling: does it occur in native cardiac tissue? J. Mol. Cell. Cardiol. 70, 25. Mahoney, V. M., Mezzano, V., Mirams, G. R., Maass, K., Li, Z.,
37–46 Cerrone, M., Vasquez, C., Bapat, A., Delmar, M., and Morley, G. E.
2. Fróes, M. M., Correia, A. H., Garcia-Abreu, J., Spray, D. C., (2016) Connexin43 contributes to electrotonic conduction across
Campos de Carvalho, A. C., and Neto, M. V. (1999) Gap-junctional scar tissue in the intact heart. Sci. Rep. 6, 26744
coupling between neurons and astrocytes in primary central nervous 26. Salvarani, N., Maguy, A., De Simone, S. A., Miragoli, M., Jousset, F.,
system cultures. Proc. Natl. Acad. Sci. USA 96, 7541–7546 and Rohr, S. (2017) TGF-b1 (transforming growth factor-b1) plays a

CARDIOMYOCYTE-MYOFIBROBLAST CX43-DEPENDENT MOVEMENT 15


g by Saab Medical Library, American Univ of Beruit (UAB) Levant U (193.188.128.21) on June 30, 2019. The FASEB Journal Vol. ${article.issue.getVolume()}, No. ${article.issue.get
pivotal role in cardiac myofibroblast arrhythmogenicity. Circ. 45. Thompson, S. A., Blazeski, A., Copeland, C. R., Cohen, D. M., Chen,
Arrhythm. Electrophysiol. 10, e004567 C. S., Reich, D. M., and Tung, L. (2014) Acute slowing of cardiac
27. Lefroy, D. C., Fang, J. C., Stevenson, L. W., Hartley, L. H., Friedman, conduction in response to myofibroblast coupling to cardiomyocytes
P. L., and Stevenson, W. G. (1998) Recipient-to-donor atrioatrial con- through N-cadherin. J. Mol. Cell. Cardiol. 68, 29–37
duction after orthotopic heart transplantation: surface electrocardio- 46. Zhang, Y., Kanter, E. M., Laing, J. G., Aprhys, C., Johns, D. C., Kardami,
graphic features and estimated prevalence. Am. J. Cardiol. 82, 444–450 E., and Yamada, K. A. (2008) Connexin43 expression levels influence
28. Roell, W., Klein, A. M., Breitbach, M., Becker, T. S., Parikh, A., Lee, J., intercellular coupling and cell proliferation of native murine cardiac
Zimmermann, K., Reining, S., Gabris, B., Ottersbach, A., Doran, R., fibroblasts. Cell Commun. Adhes. 15, 289–303
Engelbrecht, B., Schiffer, M., Kimura, K., Freitag, P., Carls, E., Geisen, 47. Mahoney, V. M., Mezzano, V., and Morley, G. E. (2016) A review of the
C., Duerr, G. D., Sasse, P., Welz, A., Pfeifer, A., Salama, G., Kotlikoff, literature on cardiac electrical activity between fibroblasts and
M., and Fleischmann, B. K. (2018) Overexpression of Cx43 in cells of myocytes. Prog. Biophys. Mol. Biol. 120, 128–133
the myocardial scar: correction of post-infarct arrhythmias through 48. Rhett, J. M., and Gourdie, R. G. (2012) The perinexus: a new feature
heterotypic cell-cell coupling. Sci. Rep. 8, 7145 of Cx43 gap junction organization. Heart Rhythm 9, 619–623
29. Thompson, S. A., Copeland, C. R., Reich, D. H., and Tung, L. (2011) 49. Asklund, T., Appelskog, I. B., Ammerpohl, O., Ekström, T. J., and
Mechanical coupling between myofibroblasts and cardiomyocytes Almqvist, P. M. (2004) Histone deacetylase inhibitor 4-phenylbutyrate
slows electric conduction in fibrotic cell monolayers. Circulation 123, modulates glial fibrillary acidic protein and connexin 43 expression,
2083–2093 and enhances gap-junction communication, in human glioblastoma
30. Zhang, P., Su, J., and Mende, U. (2012) Cross talk between cardiac cells. Eur. J. Cancer 40, 1073–1081
myocytes and fibroblasts: from multiscale investigative approaches to 50. Fu, H. Y., Sanada, S., Matsuzaki, T., Liao, Y., Okuda, K., Yamato, M.,
mechanisms and functional consequences. Am. J. Physiol. Heart Circ. Tsuchida, S., Araki, R., Asano, Y., Asanuma, H., Asakura, M., French,
Physiol. 303, H1385–H1396 B. A., Sakata, Y., Kitakaze, M., and Minamino, T. (2016) Chemical
31. Schultz, F., Hasan, A., Alvarez-Laviada, A., Miragoli, M., Bhogal, N., endoplasmic reticulum chaperone alleviates doxorubicin-induced
Wells, S., Poulet, C., Chambers, J., Williamson, C., and Gorelik, J. cardiac dysfunction. Circ. Res. 118, 798–809
(2016) The protective effect of ursodeoxycholic acid in an in vitro 51. Takatori, O., Usui, S., Okajima, M., Kaneko, S., Ootsuji, H., Takashima,
model of the human fetal heart occurs via targeting cardiac S. I., Kobayashi, D., Murai, H., Furusho, H., and Takamura, M. (2017)
fibroblasts. Prog. Biophys. Mol. Biol. 120, 149–163 Sodium 4-phenylbutyrate attenuates myocardial reperfusion injury by
32. Lyon, A. R., MacLeod, K. T., Zhang, Y., Garcia, E., Kanda, G. K., Lab, reducing the unfolded protein response. J. Cardiovasc. Pharmacol. Ther.
M. J., Korchev, Y. E., Harding, S. E., and Gorelik, J. (2009) Loss of 22, 283–292
T-tubules and other changes to surface topography in ventricular 52. Rhett, J. M., Jourdan, J., and Gourdie, R. G. (2011) Connexin 43
myocytes from failing human and rat heart. Proc. Natl. Acad. Sci. USA connexon to gap junction transition is regulated by zonula occludens-
106, 6854–6859 1. Mol. Biol. Cell 22, 1516–1528
33. Lewis, C. J., Gong, H., Brown, M. J., and Harding, S. E. (2004) 53. Aasen, T., Johnstone, S., Vidal-Brime, L., Lynn, K. S., and Koval, M.
Overexpression of b 1-adrenoceptors in adult rat ventricular myocytes (2018) Connexins: synthesis, post-translational modifications, and
enhances CGP 12177A cardiostimulation: implications for ‘putative’ trafficking in health and disease. Int. J. Mol. Sci. 19, E1296
b 4-adrenoceptor pharmacology. Br. J. Pharmacol. 141, 813–824 54. Johnstone, S. R., Billaud, M., Lohman, A. W., Taddeo, E. P., and
34. Gorelik, J., Yang, L. Q., Zhang, Y., Lab, M., Korchev, Y., and Harding, Isakson, B. E. (2012) Posttranslational modifications in connexins
S. E. (2006) A novel Z-groove index characterizing myocardial surface and pannexins. J. Membr. Biol. 245, 319–332
structure. Cardiovasc. Res. 72, 422–429 55. Pogoda, K., Kameritsch, P., Retamal, M. A., and Vega, J. L. (2016)
35. Askar, S. F., Bingen, B. O., Swildens, J., Ypey, D. L., van der Laarse, A., Regulation of gap junction channels and hemichannels by
Atsma, D. E., Zeppenfeld, K., Schalij, M. J., de Vries, A. A., and phosphorylation and redox changes: a revision. BMC Cell Biol. 17
Pijnappels, D. A. (2012) Connexin43 silencing in myofibroblasts (Suppl 1), 11
prevents arrhythmias in myocardial cultures: role of maximal diastolic 56. Lampe, P. D., TenBroek, E. M., Burt, J. M., Kurata, W. E., Johnson, R. G.,
potential. Cardiovasc. Res. 93, 434–444 and Lau, A. F. (2000) Phosphorylation of connexin43 on serine368 by
36. Majumder, R., Engels, M. C., de Vries, A. A. F., Panfilov, A. V., and protein kinase C regulates gap junctional communication. J. Cell Biol. 149,
Pijnappels, D. A. (2016) Islands of spatially discordant APD alternans 1503–1512
underlie arrhythmogenesis by promoting electrotonic dyssynchrony 57. Jiang, J., Palatinus, J. A., He, H., Iyyathuraia, J., Jordan, J., McGowan,
in models of fibrotic rat ventricular myocardium. Sci. Rep. 6, 24334 F. X., Schey, K., Bultynck, G., Zhang, Z., and Gourdie, R. G. (2016)
37. Hunter, A. W., Barker, R. J., Zhu, C., and Gourdie, R. G. (2005) Zonula Abstract 16380: phosphorylation of connexin43 at serine368 is
occludens-1 alters connexin43 gap junction size and organization by necessary for induction of cardioprotection by a connexin43
influencing channel accretion. Mol. Biol. Cell 16, 5686–5698 carboxyl-terminal mimetic peptide. Circulation 134, A16380
38. Jia, Z., Bien, H., Shiferaw, Y., and Entcheva, E. (2012) Cardiac cellular 58. Lauf, U., Giepmans, B. N. G., Lopez, P., Braconnot, S., Chen, S.-C.,
coupling and the spread of early instabilities in intracellular Ca2+. and Falk, M. M. (2002) Dynamic trafficking and delivery of
Biophys. J. 102, 1294–1302 connexons to the plasma membrane and accretion to gap junctions
39. Takens-Kwak, B. R., Jongsma, H. J., Rook, M. B., and Van Ginneken, in living cells. Proc. Natl. Acad. Sci. USA 99, 10446–10451
A. C. (1992) Mechanism of heptanol-induced uncoupling of cardiac 59. Smyth, J. W., Vogan, J. M., Buch, P. J., Zhang, S. S., Fong, T. S., Hong,
gap junctions: a perforated patch-clamp study. Am. J. Physiol. 262, T. T., and Shaw, R. M. (2012) Actin cytoskeleton rest stops regulate
C1531–C1538 anterograde traffic of connexin 43 vesicles to the plasma membrane.
40. Danon, A., Zeevi-Levin, N., Pinkovich, D. Y., Michaeli, T., Berkovich, Circ. Res. 110, 978–989
A., Flugelman, M., Eldar, Y. C., Rosen, M. R., and Binah, O. (2010) 60. Connolly, A., Trew, M. L., Smaill, B. H., Plank, G., and Bishop, M. J.
Hypoxia causes connexin 43 internalization in neonatal rat (2015) Local gradients in electrotonic loading modulate the local
ventricular myocytes. Gen. Physiol. Biophys. 29, 222–233 effective refractory period: implications for arrhythmogenesis in the
41. Rosker, C., Salvarani, N., Schmutz, S., Grand, T., and Rohr, S. (2011) infarct border zone. IEEE Trans. Biomed. Eng. 62, 2251–2259
Abolishing myofibroblast arrhythmogeneicity by pharmacological 61. Rutherford, S. L., Trew, M. L., Sands, G. B., LeGrice, I. J., and Smaill,
ablation of a-smooth muscle actin containing stress fibers. Circ. Res. B. H. (2012) High-resolution 3-dimensional reconstruction of the
109, 1120–1131 infarct border zone: impact of structural remodeling on electrical
42. Nikolaev, V. O., Moshkov, A., Lyon, A. R., Miragoli, M., Novak, P., activation. Circ. Res. 111, 301–311
Paur, H., Lohse, M. J., Korchev, Y. E., Harding, S. E., and Gorelik, J. 62. Gokhale, T. A., Asfour, H., Verma, S., Bursac, N., and Henriquez, C. S.
(2010) b2-adrenergic receptor redistribution in heart failure changes (2018) Microheterogeneity-induced conduction slowing and wave-
cAMP compartmentation. Science 327, 1653–1657 front collisions govern macroscopic conduction behavior: a compu-
43. Novak, P., Li, C., Shevchuk, A. I., Stepanyan, R., Caldwell, M., Hughes, tational and experimental study. PLOS Comput. Biol. 14, e1006276
S., Smart, T. G., Gorelik, J., Ostanin, V. P., Lab, M. J., Moss, G. W. J., 63. Hubbard, M. L., and Henriquez, C. S. (2012) Microscopic variations
Frolenkov, G. I., Klenerman, D., and Korchev, Y. E. (2009) Nanoscale in interstitial and intracellular structure modulate the distribution of
live-cell imaging using hopping probe ion conductance microscopy. conduction delays and block in cardiac tissue with source-load mis-
Nat. Methods 6, 279–281; erratum: 935 match. Europace 14 (Suppl 5), v3–v9
44. Sacan, A., Ferhatosmanoglu, H., and Coskun, H. (2008) CellTrack: an
open-source software for cell tracking and motility analysis. Bio- Received for publication December 17, 2018.
informatics 24, 1647–1649 Accepted for publication May 29, 2019.

16 Vol. 33 September 2019 The FASEB Journal x www.fasebj.org SCHULTZ ET AL.


g by Saab Medical Library, American Univ of Beruit (UAB) Levant U (193.188.128.21) on June 30, 2019. The FASEB Journal Vol. ${article.issue.getVolume()}, No. ${article.issue.get

You might also like