You are on page 1of 10

Cellular Biology

Connexin 43 Downregulation and Dephosphorylation in


Nonischemic Heart Failure Is Associated With Enhanced
Colocalized Protein Phosphatase Type 2A
Xun Ai, Steven M. Pogwizd

Abstract—In nonischemic heart failure (HF), ventricular tachycardia initiates by a nonreentrant mechanism, but there is
altered conduction (that could lead to re-entry) that could arise from changes in gap junctional proteins, especially
connexin43 (Cx43). We studied Cx43 expression and phosphorylation state in the left ventricle (LV) from an
arrhythmogenic rabbit model of nonischemic HF and from patients with HF attributable to idiopathic dilated
cardiomyopathy. We also investigated the role of protein phosphatases that dephosphorylate Cx43—PP1 and PP2A. In
HF rabbit LV, Cx43 mRNA and total protein were decreased by 29% and 34%, respectively (P⬍0.05 and P⬍0.001).
In controls, Cx43 was primarily in the phosphorylated state, but with HF there was a 64% increase in nonphosphorylated
Cx43 (Cx43-NP, normalized to total Cx43; P⬍0.05). Similar results were noted in HF rabbit myocytes (P⬍0.05) and
in human idiopathic dilated cardiomyopathy LV (P⬍0.05). We found that PP1 and PP2A colocalized with Cx43 in
rabbit LV. With HF, the level of colocalized PP2A increased ⬎2.5-fold (P⬍0.002), whereas colocalized PP1 was
unchanged. We also found intercellular coupling (assessed by Lucifer Yellow dye transfer) was markedly reduced in HF.
However, okadaic acid (10 nmol/L) reduced the amount of Cx43-NP and significantly improved cell coupling in HF.
Thus, in nonischemic HF in rabbits and humans, there is a decrease in both Cx43 expression and phosphorylation that
contributes to uncoupling. Increased levels of PP2A that colocalize with Cx43 can underlie enhanced levels of Cx43-NP
in HF. Modulation of Cx43 phosphorylation may be a potential therapeutic target to improve conduction in HF. (Circ
Res. 2005;96:54-63.)
Key Words: gap junctions 䡲 phosphorylation 䡲 phosphatases 䡲 heart failure 䡲 arrhythmia

H eart failure (HF), whether nonischemic or ischemic, is


associated with a nearly 50% incidence of sudden death,
primarily from ventricular tachycardia (VT) degenerating to
adjacent cells by providing chemical and electrical commu-
nication. They are composed of connexins, a multigene
family of conserved proteins. The relative amounts, compo-
ventricular fibrillation (VF).1 Whereas VT in nonischemic HF sition, and distribution of these connexins appear to influence
initiates primarily by a nonreentrant mechanism,2 myocardi- the conduction properties of cells.7,8 Connexin43 (Cx43) is
um from patients with idiopathic dilated cardiomyopathy the major connexin protein in ventricular myocardium, and
(IDCM) exhibits nonuniform anisotropy, slow conduction, downregulation of Cx43 (mRNA and protein) has been
and conduction block3 that could underlie reentry during the demonstrated in some experimental HF models and in the
transition from VT to VF. Conduction slowing could arise failing human heart.9 Studies in Cx43 heterozygous knockout
from decreased depolarizing currents and/or decreased gap mice, and in the canine rapid pacing HF model, support the
junctional coupling.4 However, the degree of slow conduction fact that decreased Cx43 expression per se can lead to a
and block in failing myocardium appears to be out of slowing of conduction, and that arrhythmogenesis can be
proportion to the changes in active membrane properties.5 further enhanced under pathophysiologic conditions (eg,
Moreover, LV myocytes from an animal model of nonische- myocardial ischemia) and when gap junctional alterations are
mic HF exhibit markedly decreased gap junctional conduc- heterogeneous.10 –12
tance.6 Thus, alterations in intercellular coupling involving Cx43 is a phosphoprotein that can be phosphorylated by a
cardiac gap junctions may underlie slow conduction in number of kinases13 and dephosphorylated by protein phos-
nonischemic HF. phatases such as PP1 and PP2A.14 Most phosphorylation sites
Gap junctions are specialized membrane structures consist- are serine residues on the C-terminus, although threonine and
ing of arrays of intercellular channels that directly connect tyrosine phosphorylation sites are present to a much lesser

Original received April 22, 2003; resubmission received February 20, 2004; revised resubmission received November 15, 2004; accepted November
22, 2004.
From the Department of Medicine, University of Illinois at Chicago.
Correspondence to Steven M. Pogwizd, MD, Department of Medicine, University of Illinois at Chicago, 840 S Wood St, M/C 715, Chicago, IL 60612.
E-mail spogwizd@uic.edu
© 2005 American Heart Association, Inc.
Circulation Research is available at http://www.circresaha.org DOI: 10.1161/01.RES.0000152325.07495.5a

54
Ai and Pogwizd Connexin43 Dephosphorylation in Heart Failure 55

extent.13 Whereas the Cx43 phosphorylation states mediated Western blotting was performed as previously described.22 Total
by kinases can have variable effects on gap junctional Cx43 protein (Cx43-T) was assessed using a polyclonal Cx43-T
antibody (Zymed). The nonphosphorylated isoform of Cx43 (Cx43-
communication,15,16 dephosphorylation of Cx43 (by phospha- NP) was detected by a specific monoclonal Cx43-NP antibody
tases) has been shown to decrease gap junctional communi- (Zymed).17
cation—whether assessed in neonatal rat ventricular cell pairs
with activation of endogenous phosphatases14 or in perfused Immunoconfocal Microscopy and
whole rat hearts during myocardial ischemia.17 Little is Quantitative Analysis
known about whether there is altered Cx43 phosphorylation Immunolabeling was performed with polyclonal Cx43-T and mono-
in HF; however, if so, it could contribute to altered conduc- clonal Cx43-NP antibodies described, monoclonal Cx43-T antibody
(Chemicon), and monoclonal PP1 and PP2A catalytic subunit
tion and arrhythmogenesis.18 (PP2Ac) antibodies (BD Sciences). Images were collected from a
Total cellular PP1 and PP2A activity is increased in the laser scanning confocal microscope (Carl Zeiss) and quantitatively
failing heart.19 Recent studies have shown that Cx43 interacts analyzed using National Institutes of Health image software.
with a number of proteins (eg, ZO-1 and protein kinase C) to
form a protein complex.15,20 It is conceivable that protein Phosphatase Digestion and Phosphatase Inhibition
LV homogenates were incubated with calf intestine alkaline phos-
phosphatases may also directly associate with Cx43 at gap phatase (Roche) for phosphatase digestion, and isolated myocytes
junctions to modulate its phosphorylation state, as has re- were incubated with 10 nmol/L okadaic acid (OA; Sigma) for
cently been shown for other macromolecular complexes,21 phosphatase inhibition. Changes in substrate phosphorylation status
and that the level of protein phosphatases in direct proximity were analyzed by immunoblotting with Cx43-T and Cx43-NP
antibodies.
to Cx43 may be enhanced in HF.
As such, the goals of these studies were to investigate: (1) Cosedimentation and Coimmunoprecipitation
alteration of Cx43 protein and mRNA expression; (2) change Sedimentation of PP1 and PP2A from rabbit LV homogenates was
in Cx43 phosphorylation state; (3) possible colocalization of performed using microcystin-Sepharose beads (Upstate). Immuno-
PP1 and PP2A with Cx43; (4) alteration of intercellular precipitation of Cx43-T using a specific monoclonal Cx43-T anti-
coupling; and (5) the contribution of PP1 and PP2A to Cx43 body was based on the method of Ausubel et al,24 with modifica-
tions. Immunoblotting was then performed with polyclonal and
dephosphorylation and altered cell coupling in HF. Studies monoclonal Cx43-T antibodies and monoclonal PP1 and PP2Ac
were performed in LV tissue and myocytes from an arrhyth- antibodies as described.
mogenic rabbit model of nonischemic HF in which 90% of
HF rabbits exhibit spontaneous VT and 10% die suddenly.2,22 Microinjection of Lucifer Yellow
These were validated by studies in LV tissue from patients Lucifer Yellow 4% combined with Rhodamine B dextran 1% was
microinjected into one cell of each cell pair in M199 cell culture
with end-stage HF attributable to IDCM and from nonfailing
medium in the absence or presence of OA (10 nmol/L), according to
human hearts. the method of Ruiz-Meana et al.25 Microinjection and image record-
ing were performed under a confocal microscope (Carl Zeiss).
Materials and Methods
An expanded Materials and Methods section is in the online data Statistical Analysis
supplement available at http://circres.ahajournals.org. All data were presented as means⫾SEM. Differences between two
groups were evaluated using Student t test, and P⬍0.05 was
Arrhythmogenic Rabbit Nonischemic HF Model considered to be significant.
New Zealand White rabbits underwent induction of HF by aortic
insufficiency followed by thoracic aortic constriction, as previously Results
described, until LV end-systolic dimension (LVESD) exceeded 1.20 Nonischemic HF Rabbit
cm (⬇30% increase).2,22 HF and age-matched control rabbit hearts
were rapidly excised and the LV free wall was flash-frozen in liquid
Echocardiographic Analysis
nitrogen. LV myocytes were isolated as previously described.22 The HF rabbit hearts (n⫽15) exhibited marked LV dilatation and
protocol was approved by the University of Illinois at Chicago systolic dysfunction compared with their baseline condition
Animal Studies Committee. (as well as compared to age-matched controls, n⫽8; Table,
see online supplement). With HF, LV end-diastolic dimen-
Human Heart Tissue sion (LVEDD) increased by 46% (P⬍0.001), LV end-systol-
LV tissue from failing human hearts (end-stage IDCM, n⫽8, mean ic dimension (LVESD) increased by 63% (P⬍0.001), and
ejection fraction 13⫾2%) was obtained at the time of cardiac mean fractional shortening (FS) decreased by 23%
transplantation performed at University of Illinois at Chicago or
Loyola University Hospital. LV tissue from nonfailing human hearts (P⬍0.001). Heart weight and heart-to-body weight ratio were
that were not used for transplantation for technical reasons (but with increased by 66% (P⬍0.001) and 80% (P⬍0.001), respec-
normal LV function, n⫽8) was obtained through the Regional Organ tively, in HF versus controls, whereas body weight did not
Bank of Illinois. Immediately after explantation, LV tissue was differ.
flash-frozen in liquid nitrogen. The study was approved by the
Human Studies Committees of University of Illinois at Chicago, Cx43 mRNA and Protein Expression in Control
Loyola University, and Regional Organ Bank of Illinois.
and HF Rabbit LV
Northern and Western Blot Analysis Cx43 mRNA was detected by Northern blotting as a
High-yield undegraded mRNA and protein were used in all studies. single⬇3-kb band (Figure 1A, top), and its abundance was
Synthesis of RNA probes and Northern hybridization were based on normalized to GAPDH mRNA (Figure 1A, bottom). There
methods of Srivastava et al,23 with modifications. was a 29% decrease in normalized Cx43 mRNA in HF versus
56 Circulation Research January 7/21, 2005

Cx43 Phosphorylation in Control and HF Rabbit


LV Tissue
Under physiological conditions, cardiac Cx43 is primarily
phosphorylated, but phosphorylation and dephosphorylation
can be an important pathway to regulate cell– cell communi-
cation.14 –16 The appearance of the Cx43-T bands in Western
blots in Figure 1C suggests that with HF, there is a shift from
the higher-molecular-weight phosphorylated form of Cx43 to
the lower molecular weight Cx43-NP. With the Cx43-T
Western blotting data in rabbit LV tissue (as in Figure 1C),
we were consistently able to distinguish the lower-molecular-
weight (42kD) band, which represents Cx43-NP, from the
higher-molecular-weight (43 to 46 kD) bands that represent
phosphorylated isoforms of Cx43 (Cx43-P). Based on these
data, we determined that the proportion of phosphorylated to
nonphosphorylated Cx43 changed from 88:12 (or 7.5:1) in
control to 81:19 (or 4:1) with HF (Figure 1D, left), and the
ratio of Cx43-NP to Cx43-T increased 64% (P⬍0.05; Figure
1D, middle). We also assessed changes in Cx43 phosphory-
lation state directly by using an antibody specific for
Cx43-NP that has been extensively validated.17 Immunoblot-
ting revealed that Cx43-NP (normalized to total Cx43)
increased by 66% in HF versus control rabbit LV (P⬍0.05;
Figure 1C and Figure 1D, right). This result was comparable
to the data described.
To further characterize the accumulation of Cx43-NP
isoform in HF, phosphatase digestion and immunoblotting
with the Cx43-T and Cx43-NP antibodies were performed.
Figure 1C shows a greater amount of Cx43-NP isoform in HF
rabbit LV at baseline. With alkaline phosphatase treatment,
there is a dramatic increase in the Cx43-NP signal (which
now reflects the total amount of Cx43), indicating that the
phosphorylated form of Cx43 predominates in both control
and HF, but in HF there is both less Cx43-T and a greater
Figure 1. A, Northern blot images of Cx43 (top) and GAPDH proportion of Cx43-NP.
mRNA (bottom) from control and HF rabbit LV. B, Summarized
Cx43 mRNA levels (*P⬍0.05). C, Western blots of Cx43-T (top),
Cx43-NP isoform (middle), and GAPDH (bottom) from control Cx43 Phosphorylation in Isolated Rabbit Myocytes
and HF rabbits in the absence (left, ⫺AP) and presence (right, Immunoblot data from isolated rabbit LV myocytes revealed
⫹AP) of alkaline phosphatase. D, Summarized data for Cx43 that Cx43-T protein expression (measured as the 42 to 46 kDa
expression in control and HF rabbit LV (**P⬍0.001, *P⬍0.05). At
left are the changes in Cx43-T protein expression with the rela- band) was decreased 30% in HF versus controls (P⬍0.05;
tive contributions of Cx43-NP and Cx43-P indicated. In the mid- n⫽7 and n⫽5, respectively; Figure 2A and 2B, left), with a
dle are data for the ratio of Cx43-NP isoform to Cx43-T protein similar shift from phosphorylated Cx43 to the lower-
(NP/Total) using the Cx43-T antibody. At right is the ratio of
molecular-weight Cx43-NP. As mentioned, we also deter-
Cx43-NP (using a specific Cx43 phospho-antibody) to total
Cx43 (using Cx43-T antibody). mined the proportion of phosphorylated to nonphosphorylat-
ed Cx43 and found that it changed from 86:14 (or 6:1) in
controls (n⫽7 and n⫽6, respectively; P⬍0.05; Figure 1B). control to 75:25 (or 3:1) with HF (Figure 2B, left), and the
Similar results were found when Cx43 mRNA was normal- ratio of Cx43-NP to Cx43-T increased 72% (P⬍0.05; Figure
ized to 18S ribosomal mRNA (data not shown). 2B, middle). When we used the antibody specific for Cx43-
Western blot analysis was performed in LV homogenates NP, we found that with HF, the level of Cx43-NP (normal-
from HF and age-matched control hearts, and the signal ized to total Cx43) increased by 69% (P⬍0.05; Figure 2A and
increased linearly with respect to the amount of protein Figure 2B, right), results that were comparable to those noted
loaded (Figure I in the online data supplement). The poly- in LV tissue (Figure 1D).
clonal Cx43-T antibody recognizes both phosphorylated and We confirmed these changes in Cx43 phosphorylation
Cx43-NP isoforms. As such, multiple bands (corresponding states with HF by immunofluorescent staining using confocal
to Cx43-NP and higher-molecular-weight phosphorylated microscopy. Double staining was performed in isolated rabbit
isoforms of Cx43) (Figure 1C, left) migrated at ⬇43 kDa (42 LV myocytes with Cx43-T and Cx43-NP antibodies. There
to 46 kDa). Cx43 total protein (normalized to GAPDH) was was no fluorescence signal found by staining with the
decreased 34% in HF versus controls (n⫽14 and n⫽14, secondary antibody only (negative control, data not shown).
respectively; P⬍0.001; Figure 1D, left). Figure 2C shows that Cx43 protein was localized throughout
Ai and Pogwizd Connexin43 Dephosphorylation in Heart Failure 57

Figure 2. A, Immunoblot images of Cx43-T (top), Cx43-NP isoform (middle), and GAPDH (bottom) bands from isolated LV myocytes of
control and HF rabbits. B, Summarized data for Cx43 expression in control and HF rabbit LV myocytes (*P⬍0.05). At left are the
changes in Cx43-T protein expression with the relative contributions of Cx43-NP and Cx43-P indicated. In the middle are data for the
ratio of Cx43-NP isoform to Cx43-T protein (NP/Total) using the Cx43-T antibody. At right is the ratio of Cx43-NP (using a specific
Cx43 phospho-antibody) to total Cx43 (using Cx43-T antibody). C, Double immuno-fluorescence staining with Cx43 total (green, top)
and Cx43-NP isoform (red, bottom) antibodies in rabbit LV myocytes from control and HF (scale bar⫽20 ␮m). D, Bar graph of immuno-
fluorescence density results for Cx43-T (Total) and the proportion of Cx43-NP to Cx43-T (NP/Total) (*P⬍0.05).

the myocyte membrane in distinctive punctuate patterns, analysis of the Cx43 total blot (as described), we calculated
primarily at cell ends (rather than sides). There was similar that the proportion of phosphorylated to nonphosphorylated
localization between immunolabeled Cx43-T (green) and Cx43 changed from 88:12 (or ⬇7:1) in control to 80:20 (or
Cx43-NP isoform (red) signals. Cx43-T was reduced by 28% ⬇4:1) with IDCM (Figure 3D, left), and the ratio of Cx43-NP
and Cx43-NP (normalized to Cx43-T protein) was enhanced to Cx43-T increased 65% (P⬍0.01; Figure 3D, middle).
by 71% in HF myocytes versus controls (P⬍0.05; n⫽3 and There was a 60% increase (P⬍0.05; Figure 3C and Figure
n⫽3, respectively; Figure 2D), results that were consistent 3D, right) in Cx43-NP (normalized to total Cx43) in HF by
with the immunoblotting data in rabbit LV tissue and myo- using the specific Cx43-NP antibody. All these results, along
cytes above. with phosphatase digestion data shown in Figure 3E, were
comparable to the data from HF rabbit LV described.
Cx43 Expression and Phosphorylation States in
LV From Patients With IDCM Colocalization of Protein Phosphatases With Cx43
To validate the results from HF rabbit LV, similar experi- in LV and Enhanced Colocalized Protein
ments were performed in LV tissue from nonfailing and Phosphatases With HF
failing (IDCM) human hearts. The Northern blot data re- To determine whether enhanced Cx43-NP, which was ob-
vealed that Cx43 mRNA (normalized to GAPDH) was served in HF LV, is related to the interaction of protein
reduced by 28% in IDCM versus nonfailing human LV (n⫽8 phosphatases (PP1 and PP2A) with Cx43, coimmunoprecipi-
and n⫽8, respectively; P⬍0.05; Figure 3A and 3B). tation, cosedimentation, and immunoconfocal analysis were
Immunoblotting of human LV homogenates was per- performed in control and HF rabbit LV tissue.
formed using both Cx43-T and Cx43-NP antibodies, and the To explore for a possible direct interaction of PP1 and
results were normalized to GAPDH (Figure 3C and 3D). PP2A with Cx43, PP1 and PP2A proteins were first isolated
Cx43-T was decreased by 25% in IDCM versus nonfailing from homogenates by binding to microcystin (a specific
LV (n⫽8 and n⫽8, respectively; P⬍0.05, left). From band inhibitor of PP1 and PP2A), followed by immunoblotting
58 Circulation Research January 7/21, 2005

Figure 3. A, Northern blot of Cx43 mRNA (upper) and GAPDH


mRNA (lower) from LV samples of nonfailing (NF) and IDCM
human hearts. B, Changes in Cx43 mRNA expression with Figure 4. A, Western blot images of Cx43-T (top), PP1 (middle),
IDCM versus NF (P⬍0.05). C, Immunoblot images of Cx43-T and PP2Ac (bottom) from control and HF rabbit LV homoge-
(top), Cx43-NP (middle), and GAPDH (bottom) in human NF and nates after sedimentation of PP1 and PP2A with microcystin-
IDCM LV. D, Summarized data for Cx43 expression in control Sepharose beads. B, Similar Western blot images after immuno-
and IDCM human LV (*P⬍0.05, **P⬍0.01). At left are the precipitating Cx43 with monoclonal Cx43-T antibody. C,
changes in Cx43-T expression with the relative contributions of Quantitation of the levels of PP1 and PP2Ac that coimmunopre-
Cx43-NP and Cx43-P indicated. In the middle are data for the cipitate with Cx43-T antibody (normalized to immunoprecipi-
ratio of Cx43-NP isoform to Cx43-T protein (NP/Total) using the tated Cx43; **P⬍0.01).
Cx43-T antibody. At right is the ratio of Cx43-NP (using a spe-
cific Cx43 phospho-antibody) to total Cx43 (using Cx43-T anti-
body). E, Alkaline phosphatase digestion of samples from ized to immunoprecipitated Cx43 in each individual sample)
human nonfailing and IDCM hearts. revealed that the level of PP1 was unchanged, but the level of
coimmunoprecipitated PP2Ac was increased 2.5-fold in HF
analysis. We observed a single cosedimented Cx43 reactive rabbit LV tissue versus controls (n⫽4 and n⫽4, respectively;
band at 42 to 46 kDa (Figure 4A, top), along with PP1 and P⬍0.002; Figure 4C).
PP2Ac bands (Figure 4A, middle and bottom). We further confirmed the increased colocalization of
We confirmed these findings by immunoprecipitation us- PP2Ac with Cx43 in HF by using immunofluorescence
ing a monoclonal Cx43-T antibody. Along with immunopre- staining with Cx43-NP and PP2Ac antibodies in control and
cipitated Cx43 bands (which were detected by both poly- HF LV myocytes. Figure 5 shows representative confocal
clonal and monoclonal Cx43-T antibodies, and which were images where Cx43-NP isoform (red) is present in distinctive
distinct from IgG bands), immunoblotting images revealed punctate patterns throughout the cell membrane, and it
coimmunoprecipitated PP1 and PP2Ac reactive bands (Figure colocalizes with PP2Ac (green, also distributed in cytosol)
4B). IgG alone was the negative (data not shown). Quantita- that is increased 2-fold in HF myocytes (consistent with the
tion of the coimmunoprecipitated PP1 and PP2Ac (normal- immunoprecipitation and cosedimentation data). Similar re-
Ai and Pogwizd Connexin43 Dephosphorylation in Heart Failure 59

Figure 5. Double immunofluorescence


staining of control (left) and HF rabbit
(right) isolated LV myocytes with
Cx43-NP and PP2Ac antibodies alone
(top, red; middle, green), and the overlap
(bottom, yellow). Scale bar is 20 ␮m.

sults were found for double staining using Cx43-T and the injected myocyte without passing gap junction channels
PP2Ac antibodies (data not shown). PP1 also colocalized to the recipient cell (Figure 6, bottom left). Figure 6 shows
with Cx43 at distinct membrane sites, but the amount of images of the microelectrode and control, HF, and OA-
colocalized PP1 appeared unchanged in HF (data not shown). treated HF cell pairs (injected and recipient cells), and
sequential confocal images at times 1, 38, and 214 seconds.
Alteration of Cell–Cell Communication and the Figure 7A shows representative time course data for fluores-
Role of PP2A in Accumulation of cence (normalized to plateau) in the recipient cell
Dephosphorylated Cx43 in HF (30⫻80 ␮m window; Figure 6, bottom right panel), and
To define alterations of intercellular communication in HF Figure 7B shows summarized rate constant (␶, time to reach
and it modulation by protein phosphatases, we performed ⬇63% peak) data. HF myocytes had decreased cellular
Lucifer Yellow dye transfer (microinjection) in the absence coupling, reflected by slower dye transfer with an increased ␶
or presence of the protein phosphatase inhibitor OA, at a of dye transfer of 53⫾12 seconds versus 19⫾5 seconds for
concentration (10 nmol/L) that has been shown to inhibit control (P⬍0.05; n⫽6 and n⫽5, respectively). However,
PP2A, but not PP1 (Figure 6).26,27 To delineate the injected OA-treated HF cell pairs showed improved dye transfer with
cell of the cell pairs, we also injected the high-molecular- a reduced ␶ of dye transfer (␶⫽24⫾5; P⬍0.05 versus HF and
weight (10 000) Rhodamine B dextran, which is retained in P⫽NS versus control; n⫽4). Protein phosphatase inhibition
60 Circulation Research January 7/21, 2005

Figure 6. Lucifer Yellow (LY) microinjection images of end-to-


end cell pairs (left) and subsequent LY dye transfer (right, yel-
low) from a single injected cell to a recipient cell in control, HF,
and OA-treated HF myocytes at 1, 38, and 214 seconds (right). Figure 7. A, Representative fitted single time courses of LY
A representative 30⫻80 ␮m window, which was used to quanti- transfer in recipient cell from end-to-end cell pairs of control,
tate dye transfer (fluorescence intensity in the recipient myo- HF, and 10 nmol/L OA-treated control and HF myocytes,
cyte), is shown in the bottom right panel. At bottom left is an respectively. B, Summarized ␶ data of LY transfer in recipient
example of Rhodamine B dextran (Rh, red) retained in the cell from control, HF, and OA-treated control and HF myocytes
injected myocyte. Scale bar at lower left⫽20 ␮m. (*P⬍0.05). C, Representative Western blot images of Cx43-NP
and GAPDH from myocytes in the absence or presence of OA
(10 nmol/L).
by OA (10 nmol/L) was verified by immunoblotting, with
OA-treated HF myocytes exhibiting a 42⫾9.3% decrease in increase in the level of colocalized PP2Ac in HF. Moreover,
Cx43-NP (Figure 7C; n⫽3). OA-treated control cell pairs we found that intercellular coupling between LV myocytes
showed no additional improvement in cell coupling was reduced markedly in HF. However, OA reduced the
(␶⫽18⫾7; n⫽4; P⫽NS versus untreated control cell pairs) amount of Cx43-NP and significantly improved cell coupling
(Figure 7A). in HF. Our observations suggest that both reduced Cx43
expression and enhanced Cx43-NP within gap junctions
Discussion underlie impaired cellular coupling and ultimately altered
In the present study, we studied LV tissue and myocytes from conduction in nonischemic HF. The increased levels of
an arrhythmogenic rabbit model of nonischemic HF and from colocalized PP2Ac (with Cx43) contribute to enhanced
age-matched control rabbits. We found that Cx43 expression Cx43-NP in HF.
was downregulated on both a protein and mRNA level. HF
rabbits exhibited a significant enhancement of the nonphos- Reduced Cx43 Expression and Increased Cx43-NP
phorylated isoform of Cx43 (and decreased phosphorylated Is Associated With Altered Cell Coupling and
Cx43). All these results were validated in LV tissue from Conduction in Nonischemic HF
hearts of patients with end-stage IDCM (at the time of Ventricular arrhythmias (VT and VF) account for nearly half
transplantation) compared with nonfailing hearts. We also of deaths in patients with HF (both nonischemic and ische-
observed that protein phosphatases PP1 and PP2Ac both mic).1 In 3-dimensional cardiac mapping studies, we have
colocalized with Cx43 in rabbit LV, and there was a 2.5-fold previously shown that spontaneously occurring VT in nonis-
Ai and Pogwizd Connexin43 Dephosphorylation in Heart Failure 61

chemic HF (both in our HF rabbit model and in IDCM We reasoned that local levels of an enzyme (rather than
patients) is initiated and maintained by a nonreentrant mech- total cellular levels) may best reflect its actions, and that PP1
anism such as delayed or early afterdepolarizations.2 How- and PP2A might interact directly with Cx43. Cx43 is a part of
ever, mapping in human IDCM has revealed altered aniso- a protein complex, and it has been reported that kinases and
tropic conduction and conduction block that could underlie structural proteins have direct interactions with cardiac
the development of reentry (especially during the transition Cx43.15,20 The present study provides the first evidence of an
from VT to VF).3 interaction between Cx43 and PP1 and PP2Ac in rabbit LV.
In the present study, we found that HF myocytes exhibit Cx43, PP1, and PP2A form a close complex that could be
reduced cellular coupling. This was associated with decreased immunoprecipitated by a Cx43 antibody or sedimented by
Cx43 expression on both an mRNA and protein level, similar microcystin-Sepharose beads (that bind PP1 and PP2A). We
to that reported by others.11 Cx43 downregulation may be a confirmed this with immunofluorescence staining showing
common feature of failing heart,9 although the underlying colocalization at gap junctional sites in the membrane. With
biochemical basis remains to be determined. There has been HF, we found that the level of coimmunoprecipitated PP2Ac
considerable debate as to the role of Cx43 in modulating with Cx43 was dramatically increased ⬎2.5-fold, whereas
conduction.10,29 However, studies in heterozygous Cx43 colocalized PP1 was unchanged. Moreover, OA, at a concen-
knockout mice,15 along with those in other transgenic mod- tration that inhibits PP2A, but not PP1,26,27 and that increased
els,30 and in dogs with pacing-induced HF,11 support the the levels of phosphorylated Cx43, significantly improved
contention that reduced Cx43 expression per se could con- cellular coupling between HF rabbit myocytes. The rapidity
tribute to altered cell-coupling and conduction in nonische- and near-complete recovery of coupling (even in the setting
mic HF. of reduced Cx43 protein levels) suggest that phosphorylation
Cx43 is a phosphoprotein that is predominantly phosphor- provides a dynamic means of regulation and that there may be
ylated in the control state. We confirmed this in control rabbit a steep relationship between Cx43 phosphorylation state and
and nonfailing human LV. With HF, we found a significant cellular coupling (that may plateau, as reflected by the
increase in the Cx43-NP on Western blots using an exten- unchanged coupling in OA-treated control cell pairs).
sively validated antibody specific for the Cx43-NP isoform.17 Whether dephosphorylated Cx43 per se reduces coupling
Under physiological conditions, the C-terminus of Cx43 is remains to be determined.17
extensively phosphorylated, primarily at serine sites, al-
though recent studies suggest there may be changes in Animal Model of HF
Our HF rabbit model exhibits contractile dysfunction, patho-
tyrosine phosphorylation in HF.31 The specific Cx43-NP
logic alterations, and arrhythmogenesis similar to nonische-
antibody used in the present study recognizes the nonphos-
mic human HF,2,22,28 and we have consistently validated
phorylated serine-rich segment of the Cx43 C-terminus that
biochemical and electrophysiologic findings in this model
includes putative phosphorylation sites for multiple kinases
with those in nonischemic HF in humans2,22 (including the
including protein kinase A, protein kinase C, and MAPK.15–17
results of the present study). This suggests that this arrhyth-
Our results of Cx43-NP in HF rabbits were consistent with
mogenic HF rabbit model is ideally suited for studies of
both LV tissue and isolated myocytes. Moreover, findings in
altered Cx43 expression, phosphorylation, and function.
human IDCM were nearly identical. Despite the consistency
of this finding, interpretation is limited because the extent to Limitations
which Cx43 is phosphorylated at baseline in rabbit and Our observations strongly suggest that increased levels of
human LV is not known. However, we took advantage of the colocalized PP2Ac (with Cx43) contribute to enhanced
Cx43 expression pattern on Western blots and determined the Cx43-NP and reduced cellular coupling in HF LV. Whether
extent of Cx43 phosphorylation in controls and HF. Our increased activity of other protein phosphatases, such as PP1
results in controls were consistent between rabbits and and calcineurin, or decreased activity of kinases contribute to
nonfailing humans (and similar to that noted by others17). The Cx43 dephosphorylation in HF remains unknown.14
changes in Cx43 phosphorylation with HF were substantive In addition to Cx43 downregulation and altered Cx43
and confirmed the relative changes in Cx43-NP using a phosphorylation, there could be changes in gap junction
specific Cx43-NP antibody. distribution with HF that may modulate conduction. Although
this was not assessed in the present study, recent studies have
The Role of Phosphatases in Cx43 demonstrated altered gap junction distribution in human
Dephosphorylation in Nonischemic HF IDCM.33 We focused on alterations in Cx43, by far the
Reduced Cx43 phosphorylation in HF could be attributable to predominant ventricular gap junction proteins, so we cannot
reduced phosphorylation by kinases (eg, protein kinase A, rule out alterations in the expression of other connexins (eg,
protein kinase C) and/or to increased dephosphorylation by Cx45 or Cx40) in HF.
protein phosphatases. Here, we focused on the role of
phosphatases PP1 and PP2A, the primary phosphatases that Implications
dephosphorylate Cx43,14 because levels of total cellular PP1 Our findings in the present study suggest that the combination
and total cellular PP1 and PP2A activity are increased in of Cx43 downregulation and altered phosphorylation contrib-
HF.19 Moreover, activation of phosphatases that dephosphor- ute to decreased cellular coupling and, ultimately, slow
ylate Cx43 can reduce communication between cells.14 conduction in nonischemic HF (Figure 8). The colocalization
62 Circulation Research January 7/21, 2005

ischemic heart.17 Heterogeneity of these changes in HF would


be expected to further enhance the effects on conduction.
Moreover, if Cx43 dephosphorylation enhances gap junction
degradation,32 the degree of Cx43 dephosphorylation in HF
could be underestimated, and may even contribute in part to
the decreased Cx43 protein levels.
Our findings, including the improved cellular coupling
with OA in HF, suggest that modulation of Cx43 phosphor-
ylation state and Cx43-associated proteins (eg, phosphatases)
could represent novel therapeutic approaches to improve
conduction in the failing heart.

Acknowledgments
This work was supported by an American Heart Association Estab-
lished Investigator Award (to S.M.P.) and an American Heart
Association Midwest Affiliate Postdoctoral Fellowship Award (to
X.A.). We thank Dr Eric Beyer for his valuable suggestions
regarding Lucifer Yellow dye transfer experiments and for review of
this manuscript, Dr Meiling Chen and Beth Reed for helpful
suggestions for immunoconfocal experiments, and Dr Mohammed
Islam for assistance with rabbit myocyte isolation.

References
1. Packer M. Sudden unexpected death in patients with congestive heart
failure: a second frontier. Circulation. 1985;72:681– 685.
2. Pogwizd SM. Nonreentrant mechanisms underlying spontaneous ventric-
ular arrhythmias in a model of nonischemic heart failure in rabbits.
Circulation. 1995;92:1034 –1048.
3. Anderson KP, Walker R, Urie P, Ershler PR, Lux RL, Karwandee SV.
Myocardial electrical propagation in patients with idiopathic dilated car-
diomyopathy. J Clin Invest. 1993;92:122–140.
4. Shaw RM, Rudy Y. Ionic mechanisms of propagation in cardiac tissue:
Roles of the sodium and L-type calcium currents during reduced excit-
ability and decreased gap junction coupling. Circ Res. 1997;81:727–741.
5. Vermeulen JT, McGuire MA, Opthof T, Coronel R, de Bakker JMT,
Klopping C, Janse MJ. Triggered activity and automaticity in ventricular
trabeculae of failing human and rabbit hearts. Cardiovasc Res. 1994;28:
1547–1554.
6. De Mello WC. Renin-angiotensin system and cell communication in the
failing heart. Hypertension. 1996;27:1267–1272.
7. Kumar NM, Gilula NB. The gap junction communication channel. Cell.
1996;84:381–388.
8. Saffitz JE, Davis LM, Darrow BJ, Kanter HL, Laing JG, Beyer EC. The
molecular basis of anisotropy: role of gap junctions. J Cardiovasc Elec-
trophysiol. 1995;6:498 –510.
9. Wang XJ, Gerdes AM. Chronic pressure overload cardiac hypertrophy
Figure 8. Schematic diagram showing changes in Cx43 expres- and failure in guinea pigs: III. Intercalated disc remodeling. J Mol Cell
sion, phosphorylation state, and colocalized phosphatases with Cardiol. 1999;31:333–343.
HF. With HF, the combination of decreased Cx43 expression 10. Guerrerro PA, Schuessler RB, Davis LM, Beyer EC, Johnson CM,
and enhanced nonphosphorylated Cx43 (arising from increased Yamada KA, Saffitz JE. Slow ventricular conduction in mice het-
associated PP2A) contribute to reduced cell-coupling and erozygous for a connexin43 null mutation. J Clin Invest. 1997;99:
arrhythmogenesis. 1991–1998.
11. Poelzing S, Rosenbaum DS. Altered connexin43 expression produces
arrhythmia substrate in heart failure. Am J Physiol Heart Circ Physiol.
of PP1 and PP2Ac with Cx43 indicates that Cx43 represents 2004;287:H1762–H1770.
a macromolecular complex with numerous components, in- 12. Lerner DL, Yamada KA, Schuessler RB, Saffitz JE. Accelerated onset
cluding both kinases and phosphatases. Enhanced levels of and increased incidence of ventricular arrhythmias induced by ischemia
in Cx43-deficient mice. Circulation. 2000;101:547–552.
colocalized PP2Ac in HF indicate an additional regulation of 13. Kwak BR, Jongsma H. Regulation of cardiac gap junction channel per-
gap junctional function in HF that is locally controlled. meability and conductance by several phosphorylating conditions. Mol
Alterations in Cx43 phosphorylation could modulate con- Cell Biochem. 1996;157:93–99.
14. Duthe F, Plaisance I, Sarrouilhe D, Herve JC. Endogenous protein phos-
nexin channel gating, as well as connexin trafficking and
phatase 1 runs down gap junctional communication of rat ventricular
degradation.32 Whereas effects of Cx43 phosphorylation on myocytes. Am J Physiol Cell Physiol. 2001;281:C1648 –C1656.
channel gating are complex (dependent on specific kinases 15. Bowling N, Huang X, Sandusky GE, Fouts RL, Mintze K, Esterman M,
and phosphorylation sites), decreased Cx43 phosphorylation Allen PD, Maddi R, McCall E, Vlahos CJ. Protein kinase C-␣ and -⑀
modulate connexin43 phosphorylation in human heart. J Mol Cell
has been shown to be associated with decreased gap junc-
Cardiol. 2001;33:789 –798.
tional conductance, whether assessed in vitro in cell pairs 16. Darrow BJ, Fast VG, Kleber AG, Beyer EC, Saffitz JE. Functional and
with activation of protein phosphatases14 or in vivo in the structural assessment of intercellular communication. Increased con-
Ai and Pogwizd Connexin43 Dephosphorylation in Heart Failure 63

duction velocity and enhanced connexin expression in dibutyryl cAMP- 26. Herzig S, Neuman J. Effects of serine/threonine protein phosphatases on
treated cultured cardiac myocytes. Circ Res. 1996;79:174 –183. ion channels in excitable membranes. Physiol Rev. 2000;80:173–210.
17. Beardslee MA, Lerner DL, Tadros PN, Laing JG, Beyer EC, Yamada KA, 27. Bialojan C, Takai A. Inhibitory effect of a marine-sponge toxin, okadaic
Kleber AG, Schuessler RB, Saffitz JE. Dephosphorylation and intra- acid, on protein phosphatases. Specificity and kinetics. Biochem J 1988;
cellular redistribution of ventricular connexins 43 during electrical 256:283–290.
uncoupling induced by ischemia. Circ Res. 2000;87:656 – 662. 28. Pogwizd SM, Schlotthauer K, Li L, Yuan W, Bers DM. Arrhythmo-
18. Akar FG, Spragg DD, Tunin RS, Kass DA, Tomaselli GF. Mechanisms
genesis and contractile dysfunction in heart failure: Roles of sodium-
underlying conduction slowing and arrhythmogenesis in nonischemic
calcium exchange, inward rectifier potassium current, and residual beta-
dilated cardiomyopathy. Circ Res. 2004;95:717–725.
19. Neumann J, Eschenhagen T, Jones LR, Linck B, Schmitz W, Scholz H, adrenergic responsiveness. Circ Res. 2001;88:1159 –1167.
Zimmermann N. Increased expression of cardiac phosphatases in patients 29. Morley GE, Vaidya D, Samie FH, Lo C, Delmar M, Jalife J. Character-
with end-stage heart failure. J Mol Cell Cardiol. 1997;29:265–272. ization of conduction in the ventricles of normal and heterozygous Cx43
20. Barker RJ, Price RL, Gourdie RG. Increased association of ZO-1 with knockout mice using optical mapping. J Cardiovasc Electrophysiol.
connexin43 during remodeling of cardiac gap junctions. Circ Res. 2002; 1999;10:1361–1375.
90:317–324. 30. Gutstein DE, Morley GE, Tamaddon H, Vaiday D, Schneider MD, Chen
21. Marx SO, Reiken S, Hisamatsu Y, Jayaraman T, Burkhoff D, Rosemblit N, J, Chien KR, Stuhlmann H, Fishman GI. Conduction slowing and sudden
Marks AR. PKA phosphorylation dissociates FKBP12.6 from the calcium arrhythmic death in mice with cardiac-restricted inactivation of con-
release channel: defective regulation in failing hearts. Cell. 2000;101:365–376. nexin43. Circ Res. 2001;88:333–339.
22. Pogwizd SM, Qi M, Yuan W, Samarel AM, Bers DM. Upregulation of 31. Toyofuku T, Yabuki M, Otsu K, Kuzuya T, Tada M, Hori M. Functional
Na⫹/Ca2⫹ exchanger expression and function in an arrhythmogenic rabbit role of c-Src in gap junctions of the cardiomyopathic heart. Circ Res.
model of heart failure. Circ Res. 1999;85:1009 –1019.
1999;85:672– 681.
23. Srivastava RA, Schonfeld G. Use of riboprobes for Northern blotting
32. Musil LS, Goodenough DA. Biochemical analysis of connexin43 intra-
analysis. BioTechniques. 1991;11:584 –588.
24. Ausubel FM, Brent R, Kingston RE, Moore DD, Seidman JG, Smith JA, cellular transport, phosphorylation, and assembly into gap junctional
Struhl K. Short protocol in molecular biology. 4th ed. New York: John plaques. J Cell Biol. 1991;115:1357–1374.
Wiley and Sons; 1999:90 –91. 33. Kostin S, Rieger M, Dammer S, Hein S, Richter M, Klovekorn WP,
25. Ruiz-Meana M, Garcia-Dorado D, Hofstaetter B, Piper HM, Soler-Soler Bauer ER, Schaper J. Gap junction remodeling and altered connexin43
J. Propagation of cardiomyocyte hypercontracture by passage of Na⫹ expression in the failing human heart. Mol Cell Biochem. 2003;242:
through gap junctions. Circ Res. 1999;85:280 –287. 135–144.

You might also like