ArticlePDF Available

Analysis of Connexin43 phosphorylated at S325, S328 and S330 in normoxic and ischemic heart

Authors:

Abstract and Figures

The functional consequences of Connexin43 (Cx43) phosphorylation remain largely unexplored. Using an antibody that specifically recognizes Cx43 phosphorylated at serine residues 325, 328 and/or 330 (pS325/328/330-Cx43), we show that labeling of this form of Cx43 as well as of total Cx43 is restricted to the intercalated disk region of normal ventricular tissue. In ischemic heart, significant relocalization of total Cx43 to the lateral edges of myocytes was evident; however pS325/328/330-Cx43 remained predominately at the intercalated disk. Western blots indicated a eightfold decrease in pS325/328/330-Cx43 in ischemic tissue. Peptide-binding- and competition-experiments indicated that our antibody mainly detected Cx43 phosphorylated at S328 and/or S330 in heart tissue. To evaluate how this change in Cx43 phosphorylation contributes to ischemia-induced downregulation of intercellular communication, we stably transfected Cx43(-/-) cells with a Cx43 construct in which serine residues 325, 328 and 330 had been mutated to alanine (Cx43-TM). Cx43-TM was not efficiently processed to isoforms that have been correlated with gap junction assembly. Nevertheless, Cx43-TM cells were electrically coupled, although development of coupling was delayed. Fully opened channels were only rarely observed in Cx43-TM cells, and Lucifer-Yellow-dye-coupling was significantly reduced compared with wild-type cells. These data suggest that phosphorylation of Cx43 at serine residues 325, 328 and/or 330 influences channel permselectivity and regulates the efficiency of gap junction assembly.
Content may be subject to copyright.
Analysis of Phosphorylation of Connexin43 at S325/328/330 in
Normoxic and Ischemic Heart
Paul D. Lampe
1,*
, Cynthia D. Cooper
1
, Timothy J. King
1,2
, and Janis M. Burt
3
1 Molecular Diagnostics Program, Fred Hutchinson Cancer Research Center and Department of
Pathobiology, University of Washington, Seattle, WA
3 Department of Physiology, University of Arizona, Tucson, AZ
Summary
The functional consequences of Connexin43 (Cx43) phosphorylation remain largely unexplored.
Using an antibody that specifically recognizes Cx43 phosphorylated at serines 325/328/330
(pS325/328/330), we show that this form of Cx43 as well as total Cx43 labeling was restricted to the
intercalated disk region of normal ventricular tissue. In ischemic heart, significant relocalization of
total Cx43 to the lateral edges of myocytes was evident; however pS325/328/330-Cx43 remained
predominately at the intercalated disk. Western immunoblots indicated a 8-fold decrease in
pS325/328/330-Cx43 in ischemic tissue. Peptide binding and competition experiments indicated that
our antibody mainly detected Cx43 phosphorylated at S328 and/or S330 in heart tissue. To evaluate
how this change in Cx43 phosphorylation might contribute to ischemia-induced downregulation of
intercellular communication, we stably transfected Cx43
/
cells with Cx43-S325/328/330A (Cx43-
TM). Cx43-TM was not efficiently processed to isoforms that have been correlated with gap junction
assembly. Nevertheless, Cx43-TM cells were electrically coupled, although coupling developed over
a slowed time course. Fully open channels were only rarely observed in the Cx43-TM cells, and
Lucifer Yellow dye coupling was significantly reduced compared to wild-type. These data suggest
that phosphorylation of Cx43 at serines 325, 328 and/or 330 may influence channel permselectivity
and regulate the efficiency of gap junction assembly.
Keywords
Connexin43; Gap junction; heart; ischemia; phosphorylation
Introduction
Gap junction mediated intercellular communication facilitates direct communication among
adjacent cells by allowing passage of ions and small metabolites (White and Paul, 1999; Saez
et al., 2003; Sohl and Willecke, 2004). Vertebrate gap junctions, composed of integral
membrane proteins from the Connexin gene family, are critically important in regulating
embryonic development, coordinated contraction of excitable cells, tissue homeostasis, normal
cell growth and differentiation (Saez et al., 2003; Sohl and Willecke, 2004). Furthermore,
connexin mutations have been linked to several diseases (Bergoffen et al., 1993; Gong et al.,
1997; Kelsell et al., 1997) including oculodentodigital dysplasia, a disease linked to
Connexin43 (Cx43) mutations that can cause atrioseptal defects and arrhythmias (Paznekas et
al., 2003). Twenty-one connexin genes have been identified in humans (Sohl and Willecke,
*to whom correspondence should be addressed: Paul D. Lampe, Ph.D., Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue
N., M5C800, P.O. Box 19024, Seattle, WA 98109, plampe@fhcrc.org, P: 206-667-4123, F: 206-667-2537.
2
Current Address: Hawaii Biotech, Inc., Aiea, HI
NIH Public Access
Author Manuscript
J Cell Sci. Author manuscript; available in PMC 2007 February 15.
Published in final edited form as:
J Cell Sci. 2006 August 15; 119(Pt 16): 3435–3442.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
2004). During intercellular channel formation, six connexin proteins oligomerize into a hemi-
channel or connexon; connexons are then transported to the plasma membrane by as yet
unknown mechanisms. The intact channel is formed when one hemi-channel docks with a
second in an opposing cell. Once assembled, groups of these intercellular channels (termed
gap junctional plaques) mediate the diffusion of ions, amino acids, second messengers and
other metabolites between the cytoplasms of the two cells (White and Paul, 1999; Sohl and
Willecke, 2004). The channels can be gated in response to various stimuli, including changes
in voltage, pH and connexin phosphorylation. Regulation of gap junctional communication
could occur by controlling any one of the steps mentioned above, however, many of the
regulatory mechanisms underlying these events remain elusive.
Cx43, the most ubiquitously expressed connexin, is differentially phosphorylated at a dozen
or more serine residues throughout its life cycle (Lampe and Lau, 2004). Cx43 from cultured
cells commonly demonstrates multiple electrophoretic isoforms when analyzed by SDS-
PAGE: a faster migrating form (sometimes referred to as P0 or NP) that includes the non-
phosphorylated isoform, and multiple slower migrating forms (sometimes termed P1 and P2
(Musil and Goodenough, 1991)). Following alkaline phosphatase treatment, the
phosphorylated species collapse to the fastest migrating form, suggesting that phosphorylation
is the primary covalent modification detected in SDS-PAGE analysis although no assignment
of specific phosphorylation sites to a change in Cx43 migration has been made. Pulse-chase
studies using Brefeldin A indicate some Cx43 phosphorylation occurs prior to reaching the
plasma membrane (Laird et al., 1995). This phosphorylation event may be necessary for
maintaining hemichannels in their closed state until docking occurs (Bao et al., 2004). In
addition, studies investigating phosphorylation in normal rat kidney (NRK) cells show that
Cx43 acquires resistance to Triton X-100 once it has been phosphorylated to the slower
migrating isoforms and assembled into gap junction plaques (Musil and Goodenough, 1991).
Thus, uncharacterized phosphorylation events have been correlated with changes in assembly,
acquisition of Triton X-100 insolubility and, potentially, degradation of Cx43 gap junction
channels.
In the normally functioning ventricle, Cx43 is localized to intercalated disks where it supports
the longitudinal and transverse spread of the action potential resulting in coordinated contractile
activation. Myocardial ischemia leads to Cx43 “dephosphorylation” and loss of localization to
the intercalated disk, which likely contributes to contractile failure and arrhythmias (Beardslee
et al., 2000; Schulz et al., 2003). Casein kinase 1 (CK1) mediates phosphorylation of Cx43 at
S325/328/330 in vitro. In cultured cells these sites are routinely phosphorylated; inhibition of
CK1 reduces phosphorylation at these sites and reduces gap junction assembly (Cooper and
Lampe, 2002). We sought to determine here whether phosphorylation at these sites occurs in
heart tissue and whether this phosphorylation event is affected during ischemia. Using an
antibody specific for Cx43 when it is phosphorylated at S325/328/330, we show that Cx43
localized to intercalated disks is phosphorylated at one or more of these residues (likely S328
and S330) and that ischemia leads to loss of this phosphorylation and relocalization of the
protein. Furthermore, we show that mutation of these sites increases the migration of the Cx43
phospho-isoforms on SDS-PAGE, reduces the extent of gap junction formation and reduces
gap junctional communication.
Methods
Antibodies and Reagents
All general chemicals, unless otherwise noted, were purchased from Fisher Scientific. Mouse
anti-Cx43 antibodies, Cx43CT1, Cx43CT2, and Cx43IF1 were prepared against amino acids
360–382 of Cx43 and antibody Cx43NT1 against amino acids 1–20 (Cx43NT1) of Cx43 at
the Fred Hutchinson Cancer Research Center Hybridoma Development Facility (Seattle, WA).
Lampe et al. Page 2
J Cell Sci. Author manuscript; available in PMC 2007 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
A rabbit antibody against Cx43 was purchased from Sigma (St. Louis, MO, C6219). We made
a rabbit anti-pS325/328/330-Cx43 antibody by custom commercial preparation (ProSci Inc.,
Poway, CA; 13 week schedule) against a synthetic peptide that was phosphorylated at all 3
serines (CQAG(pS)TI(pS)N(pS)HAQ-amide) that had been linked via the N-terminal cysteine
to maleimide-activated KLH (Pierce Biotechnology, Rockford, IL) according to
manufacturer’s instructions. Phosphospecific antibody was affinity purified by linking to the
phosphorylated peptide and the corresponding nonphosphorylated version to SulfoLink
Coupling Gel (Pierce) and passing the serum first over a 3 ml column prepared with the
nonphosphorylated peptide gel followed by a phosphopeptide-SulfoLink column according to
the manufacturer’s instructions. After washing, phosphospecific antibody was eluted from the
column with 5% acetic acid and fractions were neutralized with ammonium acetate. Antibody
specific for phosphorylation at S325 and S328 was affinity purified by linking peptides
phosphorylated at S325/328 and S328/330 (CGQAG(pS)TI(pS)NSH-amide and CGSTI(pS)
N(pS)HAQ-amide) to SulfoLink beads to make 1.5 ml columns of each. The polyclonal
antibody was then run first over the pS328/330 column and the unbound antibody was run over
the pS325/328 column. The pS325/328 specific antibody that bound was then eluted as
described above. All antibodies were aliquoted and stored frozen at 80°C.
Cell culture
Cx43 knockout fibroblasts lacking Cx43 (clone 23-3) or exogenously expressing wild-type
Cx43 (Cx43-WT; clone 22C-3) were provided by Dr. Erica TenBroek (University of
Minnesota). Normal Rat Kidney cells (E51) were obtained from American Tissue Type
Collection (Manassas, VA). Cells were cultured in Dulbeccos Minimal Essential Medium
(DMEM, Fisher Scientific, Pittsburgh PA) supplemented with 5–10% fetal calf serum and
antibiotics (100 U/mL penicillin G and 100μg/mL streptomycin) in a humidified 5% CO
2
environment. Cx43 with serines at 325, 328 and 330 mutated to alanine (Cx43-TM), kindly
provided in pcDNA3.1 by Dr. Steven Taffet (SUNY Health Science Center, Syracuse NY),
was subcloned into pIREShygro. To make Cx43-TM expressing fibroblasts, pIREShygro
Cx43-TM was transfected into Cx43 knockout fibroblasts (clone 23-3) using lipofectAMINE
Plus transfection reagent (GibcoBRL/Invitrogen, Carlsbad CA) according to the
manufacturer’s instructions. Cells were selected at 500μg/ml Hygromycin B and were dilution
cloned in DMEM supplemented with hygromycin and endothelial cell growth supplement
(ECGS, 5mg/mL) to facilitate selection and growth of cells. Three separate clones that stably
expressed Cx43-TM were isolated (TM-A, TM-B, TM-C).
Immunodetection of Cx43 in Heart
Mouse studies were conducted under FHCRC Institutional Animal Care and Use Committee
approval. Inbred mice (11 months of age in a FVB/N:C57BL6 background) were anaesthetized
(avertin, 0.1mL/3g body weight) and hearts were excised and placed either in ice cold PBS
(calcium and glucose-free) for 30 seconds (control group) or incubated without coronary
perfusion in non-oxygenated, glucose-free PBS for 30 minutes at 37°C. After 30 seconds
(control) or 30 minutes (“ischemic”) of incubation, hearts were longitudinally bisected and
either sonicated in Laemmli sample buffer (for Western analysis) or fixed overnight at 4°C in
10% neutral-buffered formalin (for immunohistochemistry). Although not thoroughly
characterized, the 30-minute “ischemic” period in the presence or absence of 1 mM calcium
reproduces the effects of ischemia on Cx43 electrophoretic mobility and gap junction
remodeling (see results) demonstrated in better characterized models of ischemia.
Formalin-fixed tissue was processed, sectioned, immunostained and microscopically analyzed
as previously described (King and Lampe, 2004). Briefly, tissue sections were deparaffinized,
antigen retrieved, blocked and detected utilizing rabbit anti-Cx43 (1:250, Sigma) or rabbit anti-
pS325/328/330 (1:1000). Slides were washed and incubated with a biotinylated anti-rabbit
Lampe et al. Page 3
J Cell Sci. Author manuscript; available in PMC 2007 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
secondary antibody (1:250) and detected with ABC-avidin/biotin conjugate (Vectastain,
Vector Labs, Burlingame, CA).
Immunoblotting and Immunofluorescence
Approximately the same amount of cardiac tissue for each treatment was sonicated in sample
buffer supplemented with 50mM NaF, 1mM Na
3
VO
4
, 5% β-mercaptoethanol, 1 mM PMSF
and 1x Complete protease inhibitors (Roche Molecular Biochemicals, Alameda, CA).
Insoluble material was removed by centrifugation, and the soluble fraction was separated on
SDS-10% PAGE. After immunoblotting, protein was detected with a rabbit antibody against
pS325/328/330-Cx43 and a mouse anti-Cx43 (Cx43NT). The blots were also probed with anti-
GAPDH (Ambion, Austin, TX) and anti-Vinculin (Sigma) to confirm consistent loading.
Primary antibodies were visualized with fluorescent dye-labeled secondary antibodies
[AlexaFluor 680 goat anti-rabbit (Molecular Probes, Eugene, OR) and IRDye800-conjugated
donkey anti-mouse IgG (Rockland Immunochemicals)] and directly quantified using the LI-
COR Biosciences Odyssey infrared imaging system (Lincoln, NE) and associated software
(inverted images are shown).
To test the phosphospecificity of the p325/328/330 antibody, we treated with alkaline
phosphatase or left untreated equal amounts of protein lysates from fibroblasts expressing
Cx43-WT and Cx43-TM (clone A) as previously reported (Lampe et al., 1998), ran the lysates
in SDS-PAGE and performed a Western blot with Cx43NT1 and the pS325/328/330 antibodies
on the same blot as described above. To determine the specificity of binding of the antibody,
peptides representing singly phosphorylated S325, 328 and 330 (CGQAG(pS)TISN-amide,
CAGSTI(pS)NSHA-amide, CGSTISN(pS)HAQP-amide, respectively), doubly
phosphorylated S325/328 and S328/330 (CGQAG(pS)TI(pS)NSH-amide and CGSTI(pS)N
(pS)HAQ-amide) and the triply phosphorylated immunizing peptide were synthesized and
individually incubated (at 200 pg/ml) with pS325/328/330 antibody for 30 minutes prior to
probing Western blots of whole cell lysates from mouse heart (prepared and immunodetected
as described above) using a Surf-Blot apparatus (Idea Scientific, Minneapolis, MN) for the
primary antibody incubation step. The level of antibody binding was determined as described
above by LI-COR densitometry and normalized to the signal in the absence of peptide. The
amount of antibody that can bind to the different peptides was determined by linking the 6
peptides to Reacti-Bind Maleimide-activated 96-well plates (Pierce) at saturating
concentrations. The plates were washed, blocked and incubated with pS325/328/330 antibody
or the further purified pS325/328 specific antibody in quadruplicate according to
manufacturer’s instructions and with peroxidase labeled donkey anti-rabbit secondary antibody
(Chemicon, Temecula, CA). Peroxidase levels were detected using ABTS Peroxidase substrate
(KPL, Gaithersburg, MD). The amount of antibody bound was determined by normalizing to
the total Abs410nm for the immunizing peptide and mean and standard deviation were
calculated.
To test the effect of mutation at S325/328/330, fibroblasts exogenously expressing wild-type
Cx43 or Cx43-TM were grown to 90–95% confluency and cells were harvested in 1% Triton
X-100 in PBS supplemented with 50mM NaF, 1mM Na
3
VO
4
and protease inhibitors. These
samples were separated into Triton soluble and insoluble fractions by centrifugation at
13,000g at 4°C for 10 minutes. Triton insoluble fractions (pellets) were resuspended in
Laemmli sample buffer and sonicated. Duplicate parallel cultures were lysed in 1X Laemmli
SDS-sample buffer supplemented with phosphatase/protease inhibitors and 5% β-
mercaptoethanol (whole cell lysate), followed by brief sonication. Protein assays for equal
loading, electrophoresis and immunodetection using Cx43CT2 antibody was performed as
previously described (Cooper and Lampe, 2002).
Lampe et al. Page 4
J Cell Sci. Author manuscript; available in PMC 2007 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Immunofluorescence was performed as previously described (Solan et al., 2003). Cx43 was
detected with anti-Cx43 antibody (C6219) and Alexa 594 anti-rabbit secondary antibody.
DAPI was added to visualize cellular nuclei. The coverslips were mounted onto slides with
DABCO antifade medium [25mg/ml of 1,4-Diazobicyclo-(2,2,2)octane (Sigma) diluted in
Spectroglycerol (Eastman Kodak Co.) and 10% PBS, pH 8.6] and viewed with a Nikon Diaphot
TE300 fluorescence microscope, equipped with a 40x (1.3 n.a.) oil objective and a Princeton
Instruments digital camera driven by an attached PC and Metamorph imaging software.
Dye Injection
Cx43-WT or Cx43-TM expressing cells were grown in 35mm dishes to 70–80% confluency.
Donor cells were microinjected with Lucifer yellow (1mg/ml dissolved in 0.15M LiCl) and
allowed to transfer dye for three minutes. Digital images were collected at identical camera
settings on the Nikon Diaphot TE300 fluorescence microscope described above. Following
imaging, the number of recipient cells for both conditions was quantified in a blind manner.
Junctional Conductance and Single Channel Activity
Confluent cells were trypsinized and replated at low density on glass coverslips. After 2–5h
coverslips were mounted in a custom made chamber and cells were visualized on an inverted
(Olympus IMT2) microscope equipped for phase contrast observation. Cells were bathed in
solution containing (in mM): 142.5 NaCl, 4 KCl, 1 MgCl
2
, 5 glucose, 2 sodium pyruvate, 10
HEPES, 1 BaCl
2
, 1 CaCl
2
, 15 CsCl, and 10 TEACl, pH 7.2 and osmolarity 315 mOsM.
Junctional conductance was determined using dual whole-cell voltage clamp techniques as
previously described (Cottrell et al., 2003). The pipette solution contained (in mM): 120 KCl,
14 CsCl, 3 MgCl
2
, 5 glucose, 9 HEPES, 9 EGTA, 9 TEACl, 5 Na
2
ATP, 30 KOH to adjust pH
to 7.2, 315 mOsM. Because Cx45 is sometimes detected at low levels in the parental Cx43/
cell line (Martyn et al, 1997), we first evaluated each cell pair for voltage dependent loss of
junctional conductance. Junctions displaying significant voltage dependent gating at
transjunctional voltages of 40mV (>10–15% decrease in conductance in 5–10s) were not
studied further. For voltage insensitive junctions, junctional conductance was evaluated with
10mV transjunctional pulses and single channel events were visualized with a transjunctional
driving force of 40mV following superfusion with 4mM halothane (Burt and Spray, 1989).
Event amplitudes were measured by hand for each cell pair, binned into 6.25pS bins, the binned
data from all pairs pooled, and the relative frequencies of each bin calculated and plotted. Data
were fit using Origin software as previously described (Cottrell et al., 2003).
RESULTS
Ischemia-induced “dephosphorylation” of Cx43
Previous studies have shown that the electrophoretic mobility of Cx43 isolated from ischemic
heart is increased, such that most of the protein migrates at a position that includes
dephosphorylated Cx43 so the assumption has been made that these changes in electrophoretic
mobility represent a reduction in phosphorylation at undefined sites (Beardslee et al., 2000;
Jain et al., 2003). In addition, inhibition of CK1 has been shown to reduce gap junction
assembly and to phosphorylate Cx43 in vitro at one or more of serines 325/328/330 (Cooper
and Lampe, 2002). We hypothesized that phosphorylation at sites necessary for efficient
assembly of channels might be compromised in the ischemic setting. To evaluate whether these
phosphorylation events occur in control and ischemic heart tissue, we generated a polyclonal
antibody that uniquely detects Cx43 phosphorylated at S325/328/330 (pS325/328/330–the
specificity of which is described in the next section).
In agreement with previous studies, the electrophoretic mobility of total Cx43 from ischemic
heart was increased compared to control possibly indicating unspecified “dephosphorylation”
Lampe et al. Page 5
J Cell Sci. Author manuscript; available in PMC 2007 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
event(s) had occurred (Fig. 1, each lane represents a distinct heart). When detected with the
phosphospecific antibody, we found that the pS325/328/330-Cx43 content decreased ~8 fold
(p<0.03) in ischemic hearts (6-fold when the loss of total Cx43 was taken into account).
The cellular distribution of Cx43 detected with the phosphospecific antibody and one to total
Cx43 varied in ischemic vs. normal tissue. Fig. 2 shows that in normal hearts virtually all total
Cx43 and pS325/328/330-Cx43 were localized to intercalated disks. As shown previously
(Severs et al., 2004), ischemia caused a redistribution of total Cx43 from the intercalated disk
to the lateral borders of myocytes. The pS325/328/330 signal was reduced at the intercalated
disk of ischemic heart tissue, consistent with the total Cx43 reduction, but, surprisingly,
pS325/328/330-Cx43 was undetectable at the lateral borders. If phosphorylation at these sites
promotes gap junctional functionality, then transverse conduction in the ischemic heart would
not be expected to increase (dramatically) despite increased Cx43 at lateral borders because of
the reduced phosphorylation at the S325/328/330 sites and consequent poor gap junction
functionality.
Analysis of the specificity of the pS325/328/330 antibody
To test the specificity of the pS325/328/330 antibody and to determine whether
phosphorylation at S325/328/330 directly affects protein localization and gap junction
formation, we transfected constructs encoding either wild-type Cx43 (WT) or Cx43 with serine
to alanine mutations at residues 325, 328 and 330 (Cx43-TM) into fibroblasts derived from
Cx43
/
mice and examined the properties of the gap junctions formed by these cells. We first
validated the phosphospecific nature of the antibody by examining its binding to extracts from
cells expressing Cx43-WT or Cx43-TM both before and after treatment with alkaline
phosphatase. As shown in Fig. 3A, the antibody to total Cx43 bound to 41–44 kDa bands in
all cases and revealed the classic increase in mobility to the fastest migrating position upon
alkaline phosphatase treatment. In contrast, pS325/328/330-specific antibody bound to wild-
type Cx43 only in the absence of alkaline phosphatase treatment and did not bind to the Cx43-
TM under any conditions. Furthermore, pS325/328/330 antibody appeared to bind only to the
slowest migrating isoform of Cx43; the Cx43-TM expressing cells also did not efficiently
convert Cx43 to the slower migrating phosphorylated isoforms.
To examine the specificity of the pS325/328/330 antibody and determine which sites are likely
phosphorylated in vivo, we synthesized 3 peptides with S325, 328 and 330 each singly
phosphorylated, two peptides, S325/328 and S328/330, doubly phosphorylated, and the
immunogen peptide triply phosphorylated and covalently bound these peptides to an activated
96-well plate. The p325/328/330 antibody was incubated in the well and a standard enzyme-
linked immunoabsorbent assay (ELISA) was performed (Fig. 3B, open bars). The pS328/330
peptide bound almost as much total antibody as the immunizing (triply phosphorylated) peptide
indicating that most of the antibody bound an epitope centered on these sites. The singly
phosphorylated peptides bound only a small amount of antibody. To further answer the question
of the specificity of this antibody preparation, we removed the antibody that bound to the
p328/330 peptide and purified that specific for pS325/328. Analysis of the binding of this
preparation to the 6 peptides in the ELISA format indicated that the pS325/328 purified
antibody indeed bound to mostly to the pS325/328 peptide followed by the triply
phosphorylated and the pS325 singly phosphorylated peptides (data not shown). These results
indicate that the antibody preparation is phosphospecific but polyclonal with reactivity to all
3 sites. To examine the ability of the 6 peptides to block antibody binding to Cx43, we
preincubated these peptides with the pS325/328/330 antibody prior to Western blot analysis
of heart lysates. Neither the pS325 nor pS328 singly phosphorylated peptides interfered
significantly with antibody binding to intact Cx43 leaving most of the signal remaining,
whereas the pS330 singly and all of the more extensively phosphorylated peptides did reduce
Lampe et al. Page 6
J Cell Sci. Author manuscript; available in PMC 2007 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
the Western signal to close to background levels (Fig. 3B, filled bars). Further, we tested the
ability of the pS325/328 specific antibody and found that it could bind to Cx43 prepared from
heart in an immunoblot but much less extensively than the antibody prior to the removal of the
pS328/330 reactivity (data not shown). Combined these antibody binding and competition
results indicate that this polyclonal antibody can bind to Cx43 phosphorylated at S325, S328
and S330 and that Cx43 is phosphorylated at S328 and/or S330 and to a lesser extent at S325
in heart tissue.
The role of phosphorylation at S325/328/330 in gap junction assembly
To be more quantitative about the extent of gap junction formation, we assayed the ability of
three separate Cx43-TM clones to phosphorylate Cx43 to the P2 isoform and incorporate it
into a Triton X-100 insoluble fraction, a hallmark of conventional gap junctions (Musil and
Goodenough, 1991). Like most cell lines, fibroblasts expressing wild-type Cx43 showed
several isoforms of Cx43 in the whole cell lysate and a dramatic enrichment of the P2 isoform
in the Triton insoluble fraction (Fig. 4, WT lanes). Transfectants expressing Cx43 with the TM
mutations (clones A, B, C) showed multiple differences in their Cx43 migration. In the whole
cell fraction, Cx43-TM migrated primarily at P0 with a second band between P0 and P1 and
no apparent P2. There was a significant reduction in the total amount of Triton insoluble Cx43
in clone B and C and essentially no bands that co-migrated with P2 were observed in any of
the clones. Variation in total Cx43 protein levels most likely represents clonal heterogeneity
among individual Cx43 expressing clones. Since the TM-A clone showed comparable levels
of total Cx43 expression to the Cx43-WT expressing cells, we used it for most of the subsequent
studies. We conclude that phosphorylation of Cx43 at S325, 328 and/or 330 is necessary for
Cx43 to migrate as the P2 isoform.
The localization of Cx43 in the Cx43-TM expressing clones was also examined via
immunofluorescence. In Fig. 5, we show that fibroblasts transfected with wild-type Cx43
showed both intracellular and punctate, gap junctional plaque-associated Cx43 (compare to
the lack of signal in the KO panel). In contrast, the TM mutant cell lines showed predominately
intracellular Cx43 although occasional apparent appositional labeling was observed (TM-A
clone is shown).
We next determined whether cells expressing Cx43-TM supported intercellular
communication. Parental Cx43 knockout fibroblasts (KO), wild-type Cx43 expressing
fibroblasts (WT) and the three individual Cx43-TM expressing clones A-C were microinjected
with Lucifer yellow dye and evaluated for dye transfer 3 minutes later. Digital images were
taken and the number of recipient cells was quantified. As shown in Fig. 6, the number of cells
receiving dye from donor cells was 60–85% lower in the Cx43-TM clones than for Cx43-WT
expressing cells and was comparable to the coupling observed in the parental fibroblasts that
lacked Cx43. Although Cx43 protein expression levels varied between the clones (see Fig. 4),
all 3 Cx43-TM clones including TM-A, the one that expressed similar levels of Cx43, showed
significant reductions in communication compared to the wild-type control (p<0.02). These
data suggest that phosphorylation at S325, S328 and/or S330 is critical for efficient dye transfer
in mouse fibroblasts.
We next evaluated whether junctional conductance and channel behavior differed as a
consequence of the serine to alanine mutations at the S325/328/330 sites. Based on the dye-
coupling data presented above, we expected junctional conductance to be significantly reduced
in the Cx43-TM cell clones. This was indeed the case for Cx43-TM cells plated for an
equivalent period of time as wild-type cells (2–5h) - junctional conductance in Cx43-TM cells
was 3.5±0.7 nS (n=24) vs. 6.2 ±1.1 nS (n=35, p<0.05) in Cx43-WT cells. Coupling between
Cx43-TM cells improved with time such that 24–30h after plating, conductance was 4.9±0.8
nS (n=28), a value not significantly different from Cx43-WT cells at 2–5h post plating. The
Lampe et al. Page 7
J Cell Sci. Author manuscript; available in PMC 2007 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
profile of channel conductances in the Cx43-TM cells was substantially different from that
observed in wild-type cells as shown in Fig. 7A and emphasized in the difference plot (Fig.
7B), with a severely decreased frequency for activity of fully-open channels (~110pS) and
increased frequency of 60–85pS channels.
Discussion
Previous work has established that myocardial ischemia rapidly induces uncharacterized
changes in Cx43 phosphorylation and localization, however no mechanistic connections
between specific phosphorylation events and changes in conduction or localization have been
identified. Here, we show that phosphorylation of Cx43 at serines 325, 328, and/or 330 was
drastically reduced in the ischemic heart. Further, we demonstrate that Cx43 localized to the
lateral borders of myocytes was not phosphorylated at these sites, whereas the Cx43 that
remained at the intercalated disk retained these modifications. Finally, we show that when these
sites were not phosphorylated, the event frequency for fully open channels, the overall
junctional conductance and the extent of Lucifer Yellow dye coupling were all significantly
reduced.
Although direct Cx43 phosphorylation on serines has long been correlated with gap junction
formation and function under basal conditions (Musil and Goodenough, 1991), few
mechanistic connections to specific phosphorylation events have been drawn. For instance,
phosphorylation at S364 was shown to be important for cAMP induced up-regulation of gap
junction assembly. However, cells expressing Cx43 with a S364A mutation assembled Cx43
into junctions nearly as well as wild-type expressing cells, suggesting S364 was not necessary
for assembly in homeostatic cells (TenBroek et al., 2001). Similarly, several serines are known
to be involved in mitogen-induced downregulation of communication including serines 255,
279 and 282 (MAPK) and serines 262 and 368 (protein kinase C), but these phosphorylation
events have been reported to negatively affect channel gating properties (Lampe et al., 2000;
Cottrell et al., 2003). S325, 328 and/or 330 were previously shown to be substrates for CK1
phosphorylation, a kinase implicated in the regulation of gap junction assembly (Cooper and
Lampe, 2002). Our phosphospecific antibody could react with Cx43 phosphorylated at all 3
sites in heart tissue. We probed heart lysates with 7 other Cx43 phosphospecific antibodies that
we created or are available from commercial sources and that we find to be specific for
phosphorylation at S255, S262, S279/282, S364, S368, or S372. S325/328/330 was the only
one that gave a significant decrease in phosphorylation (results not shown, several showed
little binding to heart lysate under either condition). The antibody to pS368 showed increased
binding upon hypoxia that we have investigated separately (Ek-Vitorin et al., 2006). Similar
to a previous report (Jeyaraman et al., 2003), we found (data not shown) that ischemia led to
increased binding of an anti-Cx43 antibody we prepared to a nonphosphorylated peptide
corresponding to residues 360–382 of Cx43 that appears to be identical to Zymed 13800, an
antibody prepared to a similar epitope that has been reported to bind to “dephosphorylated
Cx43” (Nagy et al., 1997).
We probed the functional consequences of these phosphorylation events by expressing Cx43
with serine to alanine conversions at all 3 sites (i.e., the TM mutant). This mutant, rather than
double or single site mutants, was selected for study for two reasons: 1) all 3 sites appear to be
phosphorylated in heart tissue and 2) the functional consequences of phosphorylation at clusters
of (phospho)serines can be misinterpreted from single site mutants because either the remaining
sites partially or fully compensate for the missing site or the remaining sites are not
phosphorylated because phosphorylation is sequential with the mutated site necessary for
subsequent phosphorylation events (as can be the case for CK1 (Flotow et al., 1990)).
Microinjected Cx43-TM cells and parental (Cx43
/
) fibroblasts were occasionally dye
coupled to 1 or 2 cells. In contrast, although electrical coupling between Cx43-TM cells was
Lampe et al. Page 8
J Cell Sci. Author manuscript; available in PMC 2007 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
reduced compared to wild-type in the early hours after plating, significant coupling was
routinely observed at 24–30 hours. This coupling was mediated by channels with conductances
of 70–90pS, a value intermediate to the fully open channel (~110pS) and the previously
reported, phosphorylation-induced 50–60pS substate (Lampe et al., 2000). These results are
consistent with at least two explanations. First, phosphorylation at S325/328/330 is necessary
for Cx43 channels to open to their fully open state. If correct, the intermediate conductance
substate of Cx43 observed in the Cx43-TM cells would have to be poorly permeated by LY to
accommodate the low levels of dye coupling observed for these cells. Interestingly, this
intermediate conductance state is commonly observed in Cx43-WT expressing rat insulinoma
cells that do not express Cx45 (Ek-Vitorin, personal communication), and the junctions formed
by these cells are poorly permeated by dye. Second, since Cx45 is expressed at low levels in
the parental fibroblasts (Martyn et al., 1997), these intermediate channels could reflect the
activity of heteromeric Cx43–Cx45 channels (Martinez et al., 2002). This seems unlikely for
two reasons, first both single channel data sets (Cx43-TM and Cx43-WT) were obtained from
cells whose macroscopic levels of coupling were comparable, and second heteromeric Cx43–
Cx45 channels are not permeated by Lucifer Yellow (Martinez et al., 2002). Regardless of
which of these possibilities is correct, the data indicate that loss of the capacity for
phosphorylation at S325, S328 and/or S330 renders the junctions inefficient at dye transfer
and less conductive.
What mechanisms might control the level of phosphorylation at S328 and/or 330 during
hypoxia? Several groups have shown that phosphatase inhibitors block the change in Cx43
electrophoretic mobility (Duthe et al., 2001; Jeyaraman et al., 2003; Turner et al., 2004). PP1
has been reported to be the main phosphatase that is active against Cx43 in cardiomyocytes
and it appears to be constitutively active (Duthe et al., 2001; Jeyaraman et al., 2003; Turner et
al., 2004). We have shown that CK1 can phosphorylate Cx43 at S325, 328, and/or 330, and
CK1 inhibition leads to loss of phosphorylated Cx43 supporting the role of this kinase in the
phosphorylation of these sites (Cooper and Lampe, 2002). However, a constant kinase/
phosphatase cycling process has been reported to occur during hypoxia with Cx43
dephosphorylation resulting from reduced cellular ATP levels without kinase or phosphatase
activity changes or Cx43 synthesis (Duthe et al., 2001; Turner et al., 2004). Given that kinase
or phosphatase localization could also be critical, the roles that kinase activity, phosphatase
activity and ATP concentration play on the control of the level of phosphorylation at
S325/328/330 during hypoxia remain to be determined.
In summary, data presented here indicate that Cx43 present in cardiac tissue is phosphorylated
at serines 325, 328 and/or 330. During ischemia, the amount of total Cx43 is reduced at the
intercalated disk and Cx43 relocates to the myocytes’ lateral edges. Cx43 at the intercalated
disk retained phosphorylation at S325/328/330 whereas Cx43 present at the lateral edges did
not. We have shown that phosphorylation at these residues was necessary to form the P2
isoform, and these residues are important in gap junction assembly/stability. Thus, the expected
net effect of ischemia would be a measured reduction in longitudinal conduction velocity, due
to decreased localization at intercalated disks (Poelzing and Rosenbaum, 2004), without a
concomitant increase in transverse conduction velocity, despite increased localization at lateral
borders. Whether reduced phosphorylation at S325/328/330 triggers the relocalization of Cx43
to the lateral edges is not known. However, the spatially specific retention of phosphorylation
at these sites at the intercalated disk and loss at the lateral edges would be expected to help
retain the anisotropy of conduction in the heart during ischemia.
Acknowledgements
These studies were supported by Grants from the National Institutes of Health: GM055632 (PDL) and HL058732
(JMB).
Lampe et al. Page 9
J Cell Sci. Author manuscript; available in PMC 2007 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
References
Bao X, Reuss L, Altenberg GA. Regulation of purified and reconstituted connexin 43 hemichannels by
protein kinase C-mediated phosphorylation of Serine 368. J Biol Chem 2004;279:20058–20066.
[PubMed: 14973142]
Beardslee MA, Lerner DL, Tadros PN, Laing JG, Beyer EC, Yamada KA, Kleber AG, Schuessler RB,
Saffitz JE. Dephosphorylation and intracellular redistribution of ventricular connexin43 during
electrical uncoupling induced by ischemia. Circulation Research 2000;87:656–662. [PubMed:
11029400]
Bergoffen J, Scherer SS, Wang S, Oronzi Scott M, Bone LJ, Paul DL, Chen K, Lensch MW, Chance PF,
Fishbeck KH. Connexin mutations in X-linked Charcot-Marie-Tooth disease. Science
1993;262:2039–2042. [PubMed: 8266101]
Burt JM, Spray DC. Volatile anesthetics block intercellular communication between neonatal rat
myocardial cells. Circ Res 1989;65:829–837. [PubMed: 2766493]
Cooper CD, Lampe PD. Casein kinase 1 regulates connexin43 gap junction assembly. J Biol Chem
2002;277:44962–44968. [PubMed: 12270943]
Cottrell GT, Lin R, Warn-Cramer BJ, Lau AF, Burt JM. Mechanism of v-Src- and mitogen-activated
protein kinase-induced reduction of gap junction communication. Am J Physiol Cell Physiol
2003;284:C511–520. [PubMed: 12388103]
Duthe F, Plaisance I, Sarrouilhe D, Herve JC. Endogenous protein phosphatase 1 runs down gap junctional
communication of rat ventricular myocytes. American Journal of Physiology - Cell Physiology
2001;281:C1648–1656. [PubMed: 11600429]
Ek-Vitorin JF, King TJ, Heyman NS, Lampe PD, Burt JM. Selectivity of Connexin 43 Channels Is
Regulated Through Protein Kinase C-Dependent Phosphorylation. Circ Res. 2006
Flotow H, Graves PR, Wang AQ, Fiol CJ, Roeske RW, Roach PJ. Phosphate groups as substrate
determinants for casein kinase I action. J Biol Chem 1990;265:14264–14269. [PubMed: 2117608]
Gong X, Li E, Klier G, Huang Q, Wu Y, Lei H, Kumar NM, Horwitz J, Gilula NB. Disruption of alpha3
connexin gene leads to proteolysis and cataractogenesis in mice. Cell 1997;91:833–843. [PubMed:
9413992]
Jain SK, Schuessler RB, Saffitz JE. Mechanisms of delayed electrical uncoupling induced by ischemic
preconditioning. Circ Res 2003;92:1138–1144. [PubMed: 12730093]
Jeyaraman M, Tanguy S, Fandrich RR, Lukas A, Kardami E. Ischemia-induced dephosphorylation of
cardiomyocyte connexin-43 is reduced by okadaic acid and calyculin A but not fostriecin. Mol Cell
Biochem 2003;242:129–134. [PubMed: 12619875]
Kelsell DP, Dunlop J, Stevens HP, Lench NJ, Laing JN, Parry G, Mueller RF, Leigh IM. Connexin 26
mutations in hereditary non-syndromic sensorineural deafness. Nature 1997;387:80–83. [PubMed:
9139825]
King TJ, Lampe PD. The gap junction protein connexin32 is a mouse lung tumor suppressor. Cancer Res
2004;64:7191–7196. [PubMed: 15492231]
Laird DL, Castillo M, Kasprzak L. Gap junction turnover, intracellular trafficking, and phosphorylation
of connexin43 in Brefeldin A-treated rat mammary tumor cells. J Cell Biol 1995;131:1193–1203.
[PubMed: 8522583]
Int J of Biochem Cell Biol. 36. 2004. The effects of connexin phosphorylation on gap junctional
communication.
Lampe PD, Kurata WE, Warn-Cramer B, Lau AF. Formation of a distinct connexin43 phosphoisoform
in mitotic cells is dependent upon p34
cdc2
kinase. J Cell Sci 1998;111:833–841. [PubMed: 9472011]
Lampe PD, TenBroek EM, Burt JM, Kurata WE, Johnson RG, Lau AF. Phosphorylation of connexin43
on serine368 by protein kinase C regulates gap junctional communication. J Cell Biol
2000;126:1503–1512. [PubMed: 10871288]
Martinez AD, Hayrapetyan V, Moreno AP, Beyer EC. Connexin43 and connexin45 form heteromeric
gap junction channels in which individual components determine permeability and regulation. Circ
Res 2002;90:1100–1107. [PubMed: 12039800]
Lampe et al. Page 10
J Cell Sci. Author manuscript; available in PMC 2007 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Martyn KD, Kurata WE, Warn-Cramer BJ, Burt JM, TenBroek E, Lau AF. Immortalized connexin43
knockout cell lines display a subset of biological properties associated with the transformed
phenotype. Cell Growth & Differentiation 1997;8:1015–1027. [PubMed: 9300183]
Musil LS, Goodenough DA. Biochemical analysis of connexin43 intracellular transport, phosphorylation
and assembly into gap junctional plaques. J Cell Biol 1991;115:1357–1374. [PubMed: 1659577]
Nagy JI, Li WEI, Roy C, Doble BW, Gilchrist JS, Kardami E, Hertzberg EL. Selective monoclonal
antibody recognition and cellular localization of an unphosporylated form of connexin43. Exper Cell
Res 1997;236:127–136. [PubMed: 9344592]
Paznekas WA, Boyadjiev SA, Shapiro RE, Daniels O, Wollnik B, Keegan CE, Innis JW, Dinulos MB,
Christian C, Hannibal MC, et al. Connexin 43 (GJA1) mutations cause the pleiotropic phenotype of
oculodentodigital dysplasia. Am J Hum Genet 2003;72:408–418. [PubMed: 12457340]
Poelzing S, Rosenbaum DS. Nature, significance, and mechanisms of electrical heterogeneities in
ventricle. Anat Rec A Discov Mol Cell Evol Biol 2004;280:1010–1017. [PubMed: 15368342]
Saez JC, Berthoud VM, Branes MC, Martinez AD, Beyer EC. Plasma membrane channels formed by
connexins: their regulation and functions. Physiol Rev 2003;83:1359–1400. [PubMed: 14506308]
Schulz R, Gres P, Skyschally A, Duschin A, Belosjorow S, Konietzka I, Heusch G. Ischemic
preconditioning preserves connexin 43 phosphorylation during sustained ischemia in pig hearts in
vivo. Faseb J 2003;17:1355–1357. [PubMed: 12759340]
Severs NJ, Dupont E, Coppen SR, Halliday D, Inett E, Baylis D, Rothery S. Remodelling of gap junctions
and connexin expression in heart disease. Biochim Biophys Acta 2004;1662:138–148. [PubMed:
15033584]
Sohl G, Willecke K. Gap junctions and the connexin protein family. Cardiovasc Res 2004;62:228–232.
[PubMed: 15094343]
Solan JL, Fry MD, TenBroek EM, Lampe PD. Connexin43 phosphorylation at S368 is acute during S
and G2/M and in response to protein kinase C activation. J Cell Sci 2003;116:2203–2211. [PubMed:
12697837]
TenBroek EM, Lampe PD, Solan JL, Reynhout JK, Johnson RG. Ser364 of connexin43 and the
upregulation of gap junction assembly by cAMP. J Cell Biol 2001;155:1307–1318. [PubMed:
11756479]
Turner MS, Haywood GA, Andreka P, You L, Martin PE, Evans WH, Webster KA, Bishopric NH.
Reversible connexin 43 dephosphorylation during hypoxia and reoxygenation is linked to cellular
ATP levels. Circ Res 2004;95:726–733. [PubMed: 15358666]
White T, Paul D. Genetic diseases and gene knockouts reveal diverse connexin functions. Ann Rev
Physiology 1999;61:283–310.
Lampe et al. Page 11
J Cell Sci. Author manuscript; available in PMC 2007 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 1.
Increased electrophoretic mobility of Cx43 in the ischemic heart involves dephosphorylation
at serines 325/328/330. Western blot of total protein isolated from 3 control (Lanes 1,3,5,
respectively) or 3 globally ischemic (30’ at 37°C; Lanes 2,4,6, respectively) hearts
simultaneously probed for total Cx43 (Cx43NT-1) and Cx43 phosphorylated at S325/328/330
followed by GAPDH, and vinculin antibodies. Note the increased mobility of Cx43 and
decreased pS325/328/330 content in the ischemic samples. Molecular weight standards are 28,
49, 62 and 98 kDa.
Lampe et al. Page 12
J Cell Sci. Author manuscript; available in PMC 2007 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 2.
Differential phosphorylation of Cx43 localized at intercalated disks vs. lateral edges of
myocytes in the normal and ischemic heart. The localization of total Cx43 (Sigma C6219) and
pS325/328/330-Cx43 in normal and ischemic heart is shown. Note the increase in Cx43 at the
lateral edges of the myocytes of the ischemic heart. Lateral edge Cx43 was not detectably
phosphorylated at S325/328/330. In contrast, Cx43 at the intercalated disks of ischemic heart
retained phosphorylation at S325/328/330 (bar=20μm).
Lampe et al. Page 13
J Cell Sci. Author manuscript; available in PMC 2007 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 3.
The pS325/328/330 antibody is specific for the P2 isoform of Cx43 and pS328/330. (A) Equal
amounts of protein lysate from fibroblasts expressing wild-type Cx43 (WT) and Cx43-TM
(TM, clone A) were either treated with alkaline phosphatase (+AP) or untreated and probed in
a Western blot with Cx43NT1 (Total Cx43-left panel) and the pS325/328/330 (right panel)
antibodies on the same blot. Molecular weight standards are marked on the left side and the
P2 isoform is marked with an *. (B) The amount of antibody bound to the different peptides
representing singly, doubly and triply phosphorylated Cx43 linked to an ELISA well (open
bars) is shown along with the ability of these peptides to compete with Cx43 present in heart
lysates in a Western immunoblot format (filled bars). Error bars represent standard deviation
from the mean.
Lampe et al. Page 14
J Cell Sci. Author manuscript; available in PMC 2007 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 4.
Cx43-TM expressing cells do not phosphorylate Cx43 to the P2 isoform and contain less Triton
insoluble (TI) Cx43. Whole cell (WC) and Triton X-100 insoluble (TI) cellular lysates from
cells expressing wild-type Cx43 (WT) or three Cx43-TM clones (TM-A, TM-B, and TM-C)
were western immunoblotted and probed for total Cx43 with the Cx43NT1 antibody. A darker
exposure of the TM-C clone is shown. The migration position of a 50 and 36 kDa marker are
indicated with an * and the vinculin loading controls are indicated with a V. The Triton
insoluble vinculin could be associated with the cytoskeleton or adhesive junctions.
Lampe et al. Page 15
J Cell Sci. Author manuscript; available in PMC 2007 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 5.
Comparison of Cx43 localization in parental knockout (KO) cells, wild-type Cx43 (WT) and
Cx43-TM (TM-A) expressing cells. Apparent gap junctions are marked by arrowheads in the
WT panel. Bar=10μm.
Lampe et al. Page 16
J Cell Sci. Author manuscript; available in PMC 2007 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 6.
Cells expressing Cx43 S325/328/330A are inefficient at dye transfer. Wild-type (WT), parental
(KO) and Cx43-TM (clones TM-A, TM-B and TM-C) expressing cells were microinjected
with Lucifer yellow dye. After 3 minutes of transfer, digital images were taken and the number
of recipient cells was determined (n=the number of injected cells). Mean and standard error
are shown. The extent of dye transfer was significantly different (p<0.02) for WT compared
to all of the other cell types as indicated by **.
Lampe et al. Page 17
J Cell Sci. Author manuscript; available in PMC 2007 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Fig. 7.
Histogram of channel events observed in Cx43-TM (gray) and Cx43-WT expressing cells.
Panel A shows event frequency as a percent of total events in each bin; panel B shows the
difference induced by mutation of the 325, 328, and 330 sites. Note the reduced incidence of
events corresponding to fully open channels (106 ± 1 pS) and increased incidence of 75 ± 2
pS. The 54 ± 1 pS population did not differ between wild-type and mutant data sets. Difference
data were fit using Origin, Chi
2
and R
2
for fit are 5.677 and 0.94, respectively (WT: 1851
events, n=13; TM: 418 events, n=7).
Lampe et al. Page 18
J Cell Sci. Author manuscript; available in PMC 2007 February 15.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
... In some cases, posttranslational modifications of Cx43 itself may be involved. Acute cardiac ischemia led to lateralization and coincided with reduced Cx43 phosphorylation at one or more of the serines S325/328/330 (182,191); while dephosphorylated Cx43 was found both at IDs and lateral membranes, the remaining Cx43 phosphorylated at S325/328/330 localized exclusively to the ID (182). The importance of the S325/328/330 triplet was further established in KI mice of phosphomimic (S3E) or dephosphomimic (S3A) mutations in Cx43. ...
... In some cases, posttranslational modifications of Cx43 itself may be involved. Acute cardiac ischemia led to lateralization and coincided with reduced Cx43 phosphorylation at one or more of the serines S325/328/330 (182,191); while dephosphorylated Cx43 was found both at IDs and lateral membranes, the remaining Cx43 phosphorylated at S325/328/330 localized exclusively to the ID (182). The importance of the S325/328/330 triplet was further established in KI mice of phosphomimic (S3E) or dephosphomimic (S3A) mutations in Cx43. ...
... The process is believed to occur by internalization of whole plaques or of parts from the plaque interior (161,200). The internalization may be triggered by changes in phosphorylation state (182) as well as by ubiquitination (204,207). ...
Article
Full-text available
The intercalated disc (ID) is a highly specialized structure, connecting cardiomyocytes via mechanical and electrical junctions. Although described in some detail by light microscopy in the 19th century, it was in 1966 that electron microscopy images showed that the ID represented apposing cell borders, and provided detailed insight into the complex ID nanostructure. Since then, much has been learned about the ID and its molecular composition, and it has become evident that a large number of proteins, not all of them involved in direct cell-to-cell coupling via mechanical or gap junctions, reside at the ID. Furthermore, an increasing number of functional interactions between ID components are emerging, leading to the concept that the ID is not the sum of isolated molecular silos, but an interacting molecular complex, an "organelle" where components work in concert to bring about electrical and mechanical synchrony. The aim of the present review is to give a short historical account of the ID's discovery, an updated overview of its composition and organization, followed by a discussion of the physiological implications of the ID architecture and the local intermolecular interactions. The latter will focus on both the importance for normal conduction of cardiac action potentials as well as the impact on the pathophysiology of arrhythmias.
... Phosphorylation events play a particularly important role in Cx43 GJ assembly, gating, and degradation, and many of the 21 serine (S) residues in the carboxy terminus of Cx43 have been reported to be the phosphorylation targets of various kinases, including protein kinase B (PKB or Akt), protein kinase C (PKC), mitogen-activated protein kinase (MAPK), and casein kinase 1 (CK1) [28]. Several studies have shown that acute stressors including ischemia promote the internalization of Cx43 GJs by modifying the phosphorylation status of Cx43 [38][39][40], reducing cell-cell communication. In adult ventricular cardiomyocytes, ischemia mediates a decrease in both total and CK1-phosphorylated forms of Cx43, resulting in decreased sarcolemmal Cx43 and impaired electrical coupling between neighboring myocytes [40]. ...
... Several studies have shown that acute stressors including ischemia promote the internalization of Cx43 GJs by modifying the phosphorylation status of Cx43 [38][39][40], reducing cell-cell communication. In adult ventricular cardiomyocytes, ischemia mediates a decrease in both total and CK1-phosphorylated forms of Cx43, resulting in decreased sarcolemmal Cx43 and impaired electrical coupling between neighboring myocytes [40]. In an intriguing study, Remo and colleagues focused on the role of CK1 phosphorylation in cardiac Cx43 GJ remodeling and created transgenic mice in which three Cx43 CK1 phosphorylation sites (S325/328/330) were replaced by phosphomimetic glutamic acids (hereafter abbreviated as Cx43-S3E) [41]. ...
Article
Full-text available
The transplantation of human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) has shown promise in preclinical models of myocardial infarction, but graft myocardium exhibits incomplete host–graft electromechanical integration and a propensity for pro-arrhythmic behavior. Perhaps contributing to this situation, hPSC-CM grafts show low expression of connexin 43 (Cx43), the major gap junction (GJ) protein, in ventricular myocardia. We hypothesized that Cx43 expression and function could be rescued by engineering Cx43 in hPSC-CMs with a series of phosphatase-resistant mutations at three casein kinase 1 phosphorylation sites (Cx43-S3E) that have been previously reported to stabilize Cx43 GJs and reduce arrhythmias in transgenic mice. However, contrary to our predictions, transgenic Cx43-S3E hPSC-CMs exhibited reduced Cx43 expression relative to wild-type cells, both at baseline and following ischemic challenge. Cx43-S3E hPSC-CMs showed correspondingly slower conduction velocities, increased automaticity, and differential expression of other connexin isoforms and various genes involved in cardiac excitation–contraction coupling. Cx43-S3E hPSC-CMs also had phosphorylation marks associated with Cx43 GJ internalization, a finding that may account for their impaired GJ localization. Taken collectively, our data indicate that the Cx43-S3E mutation behaves differently in hPSC-CMs than in adult mouse ventricular myocytes and that multiple biological factors likely need to be addressed synchronously to ensure proper Cx43 expression, localization, and function.
... In this context, cardiac fibroblasts may display a "dome-like configuration" in their action potentials, referencing the unique shape observed in specific cardiac cell types, particularly ventricular cardiomyocytes [13,[25][26][27] (Figure 2). ...
Article
Full-text available
This review paper delves into the current body of evidence, offering a thorough analysis of the impact of large-conductance Ca2+-activated K+ (BKCa or BK) channels on the electrical dynamics of the heart. Alterations in the activity of BKCa channels, responsible for the generation of the overall magnitude of Ca2+-activated K+ current at the whole-cell level, occur through allosteric mechanisms. The collaborative interplay between membrane depolarization and heightened intracellular Ca2+ ion concentrations collectively contribute to the activation of BKCa channels. Although fully developed mammalian cardiac cells do not exhibit functional expression of these ion channels, evidence suggests their presence in cardiac fibroblasts that surround and potentially establish close connections with neighboring cardiac cells. When cardiac cells form close associations with fibroblasts, the high single-ion conductance of these channels, approximately ranging from 150 to 250 pS, can result in the random depolarization of the adjacent cardiac cell membranes. While cardiac fibroblasts are typically electrically non-excitable, their prevalence within heart tissue increases, particularly in the context of aging myocardial infarction or atrial fibrillation. This augmented presence of BKCa channels’ conductance holds the potential to amplify the excitability of cardiac cell membranes through effective electrical coupling between fibroblasts and cardiomyocytes. In this scenario, this heightened excitability may contribute to the onset of cardiac arrhythmias. Moreover, it is worth noting that the substances influencing the activity of these BKCa channels might influence cardiac electrical activity as well. Taken together, the BKCa channel activity residing in cardiac fibroblasts may contribute to cardiac electrical function occurring in vivo.
... Cardiac Cx43 exists in myocardial gap junctions, which are primarily localized in intercalated discs [16,17]. In healthy myocardium, gap junctional Cx43 is in a phosphorylated state to enhance normal intercellular communication, which enables the exchange of electrical stimuli and synchronous myocardial contraction [18][19][20]. Sufficient evidence has been Int. J. Mol. ...
Article
Full-text available
Contraction band necrosis (CBN) is a common abnormality found in the myocardium of cocaine abusers, but is rarely reported in experimental models of cocaine abuse. Connexin 43 (Cx43) is essential for cardiac intercellular communication and the propagation of CBN. Under stress or injury, cardiac Cx43 is dephosphorylated, which is related to cardiomyocyte dysfunction and pathogenesis, whereas adiponectin exerts beneficial effects in the myocardium. In this study, we explore the effects of cocaine on cardiac Cx43 in vivo. Rats were administered cocaine via the tail vein at 20 mg/kg/day for 14 days, and showed widespread CBN, microfocal myocarditis and myocardial fibrosis, corresponding to a dysfunction of cardiac mitochondria under increased oxidative stress. The increase in dephosphorylated cardiac Cx43 and its negative correlation with the myocardial distribution of CBN after cocaine administration were determined. In addition, apoptosis and necroptosis, as well as increased adiponectin levels, were observed in the myocardium after cocaine exposure. Accordingly, we found altered profiles of cardiac Cx43, CBN and its negative correlation with dephosphorylated cardiac Cx43, and the possible involvement of adiponectin in the myocardium after 14 days of cocaine administration. The latter might play a protective role in the cardiotoxicity of cocaine. The current findings would be beneficial for establishing novel therapeutic strategies in cocaine-induced cardiac consequences.
Article
Intrahepatic cholangiocarcinoma (ICC) is an aggressive bile duct malignancy that frequently exhibits isocitrate dehydrogenase ( IDH1/IDH2 ) mutations. Mutant IDH (IDHm) ICC is dependent on SRC kinase for growth and survival and is hypersensitive to inhibition by dasatinib, but the molecular mechanism underlying this sensitivity is unclear. We found that dasatinib reduced p70 S6 kinase (S6K) and ribosomal protein S6 (S6), leading to substantial reductions in cell size and de novo protein synthesis. Using an unbiased phosphoproteomic screen, we identified membrane-associated guanylate kinase, WW, and PDZ domain containing 1 (MAGI1) as an SRC substrate in IDHm ICC. Biochemical and functional assays further showed that SRC inhibits a latent tumor-suppressing function of the MAGI1–protein phosphatase 2A (PP2A) complex to activate S6K/S6 signaling in IDHm ICC. Inhibiting SRC led to activation and increased access of PP2A to dephosphorylate S6K, resulting in cell death. Evidence from patient tissue and cell line models revealed that both intrinsic and extrinsic resistance to dasatinib is due to increased phospho-S6 (pS6). To block pS6, we paired dasatinib with the S6K/AKT inhibitor M2698, which led to a marked reduction in pS6 in IDHm ICC cell lines and patient-derived organoids in vitro and substantial growth inhibition in ICC patient-derived xenografts in vivo. Together, these results elucidated the mechanism of action of dasatinib in IDHm ICC, revealed a signaling complex regulating S6K phosphorylation independent of mTOR, suggested markers for dasatinib sensitivity, and described a combination therapy for IDHm ICC that may be actionable in the clinic.
Article
Objectives: Particulate Matter (PM) air pollution is known to exacerbate cardiopulmonary diseases. We previously demonstrated that PM mediates endothelial injury and barrier disruption by modulating the endothelial cytoskeleton and cell-cell junctions, but the effects of PM exposure on cell-cell communication and gap junction activity are still unknown. Methods: This study focused on the characterization of PM-regulated endothelial dysfunction through connexin 43 (Cx43), the most abundant gap junction protein expressed in lung endothelial cells (ECs), using cultured human lung endothelial cells and a well-characterized PM sample. Results: PM exposure induced a time-dependent increase of Cx43 in human lung ECs at both the mRNA and protein levels. N-acetylcysteine (NAC), a reactive oxygen species (ROS) scavenger, significantly suppressed PM-induced Cx43 expression. Cx43 proteins on the plasma membrane and ER/Golgi apparatus were elevated in response to a PM challenge. In addition, PM induced gap junction activity, which was indicated by green fluorescence dye transfer between two adjacent ECs. Moreover, GAP27, a selective Cx43 channel inhibitor, attenuated PM-induced human lung EC barrier disruption, which was reflected by rescued trans-endothelial electrical resistance (TER) with an electric cell-substrate impedance sensing system. Moreover, knocking down Cx43 alleviated PM-induced myosin light chain (MLC) phosphorylation. Conclusions: These results strongly suggest that Cx43 plays a key role in PM-mediated endothelial barrier disruption and signal transduction. Cx43 may be a therapeutic target in PM-mediated cardiopulmonary disorders.
Article
Background: Heart failure causes changes in Cx43 (Connexin43) regulation that are associated with arrhythmic heart disease. Pyk2 (proline-rich tyrosine kinase 2) is activated in cardiomyopathies and phosphorylates Cx43 to decrease intercellular communication. This study was designed to determine if Pyk2 inhibition improves cardiac function in a myocardial infarction (MI)-induced heart failure model in rats. Methods: MI (ligation of left anterior descending artery) rats were treated with the Pyk2 inhibitor PF4618433. Hemodynamic and structural parameters were monitored in Sham (n=5), MI-vehicle (n=5), and MI-PF4618433 (n=8) groups. Heart tissues were collected after 6 weeks to assess Pyk2 and Cx43 protein level and localization. Results: PF4618433 produced no observed adverse effects and inhibited ventricular Pyk2. PF4618433 reduced the MI infarct size from 34% to 17% (P=0.007). PF4618433 improved stroke volume (P=0.031) and cardiac output (P=0.009) in comparison to MI-vehicle with values similar to the Sham group. PF4618433 also led to an increase in the ejection fraction (P=0.002) and fractional shortening (P=0.006) when compared with the MI-vehicle (32% and 35% improvement, respectively) yet were lower in comparison with the Sham group. Pyk2 inhibition decreased Cx43 tyrosine phosphorylation (P=0.043) and maintained Cx43 at the intercalated disc in the distal ventricle 6 weeks post-MI. Conclusions: Unlike other attempts to decrease Cx43 remodeling after MI-induced heart failure, inhibition of Pyk2 activity maintained Cx43 at the intercalated disc. This may have aided in the reduced infarct size (acute time frame) and improved cardiac function (chronic time frame). Additionally, we provide evidence that Pyk2 is activated following MI in human left ventricle, implicating a novel potential target for therapy in patients with heart failure.
Article
Full-text available
Mitochondrial dysfunction induced by acute cardiac ischemia–reperfusion (IR), may increase susceptibility to arrhythmias by perturbing energetics, oxidative stress production and calcium homeostasis. Although changes in mitochondrial morphology are known to impact on mitochondrial function, their role in cardiac arrhythmogenesis is not known. To assess action potential duration (APD) in cardiomyocytes from the Mitofusins-1/2 (Mfn1/Mfn2)-double-knockout (Mfn-DKO) compared to wild-type (WT) mice, optical-electrophysiology was conducted. To measure conduction velocity (CV) in atrial and ventricular tissue from the Mfn-DKO and WT mice, at both baseline and following simulated acute IR, multi-electrode array (MEA) was employed. Intracellular localization of connexin-43 (Cx43) at baseline was evaluated by immunohistochemistry, while Cx-43 phosphorylation was assessed by Western-blotting. Mfn-DKO cardiomyocytes demonstrated an increased APD. At baseline, CV was significantly lower in the left ventricle of the Mfn-DKO mice. CV decreased with simulated-ischemia and returned to baseline levels during simulated-reperfusion in WT but not in atria of Mfn-DKO mice. Mfn-DKO hearts displayed increased Cx43 lateralization, although phosphorylation of Cx43 at Ser-368 did not differ. In summary, Mfn-DKO mice have increased APD and reduced CV at baseline and impaired alterations in CV following cardiac IR. These findings were associated with increased Cx43 lateralization, suggesting that the mitofusins may impact on post-MI cardiac-arrhythmogenesis.
Article
Full-text available
We previously demonstrated that the gap junction protein connexin43 is translated as a 42-kD protein (connexin43-NP) that is efficiently phosphorylated to a 46,000-Mr species (connexin43-P2) in gap junctional communication-competent, but not in communication-deficient, cells. In this study, we used a combination of metabolic radiolabeling and immunoprecipitation to investigate the assembly of connexin43 into gap junctions and the relationship of this event to phosphorylation of connexin43. Examination of the detergent solubility of connexin43 in communication-competent NRK cells revealed that processing of connexin43 to the P2 form was accompanied by acquisition of resistance to solubilization in 1% Triton X-100. Immunohistochemical localization of connexin43 in Triton-extracted NRK cells demonstrated that connexin43-P2 (Triton-insoluble) was concentrated in gap junctional plaques, whereas connexin43-NP (Triton-soluble) was predominantly intracellular. Using either a 20 degrees C intracellular transport block or cell-surface protein biotinylation, we determined that connexin43 was transported to the plasma membrane in the Triton-soluble connexin43-NP form. Cell-surface biotinylated connexin43-NP was processed to Triton-insoluble connexin43-P2 at 37 degrees C. Connexin43-NP was also transported to the plasma membrane in communication defective, gap junction-deficient S180 and L929 cells but was not processed to Triton-insoluble connexin43-P2. Taken together, these results demonstrate that gap junction assembly is regulated after arrival of connexin43 at the plasma membrane and is temporally associated with acquisition of insolubility in Triton X-100 and phosphorylation to the connexin43-P2 form.
Article
Full-text available
Phosphorylation of rabbit muscle glycogen synthase by cyclic AMP-dependent protein kinase has been shown to enhance subsequent phosphorylation by casein kinase I (Flotow, H., and Roach, P. J. (1989) J. Biol. Chem. 264, 9126-9128). In the present study, synthetic peptides based on the sequences of the four phosphorylated regions in muscle glycogen synthase were used to probe the role of substrate phosphorylation in casein kinase I action. With all four peptides, prior phosphorylation significantly stimulated phosphorylation by casein kinase I. A series of peptides was synthesized based on the NH2-terminal glycogen synthase sequence PLSRTLS7VSS10LPGL, in which phosphorylation at Ser7 is required for modification of Ser10 by casein kinase I. The spacing between the P-Ser and the acceptor Ser was varied to have 1, 2, or 3 intervening residues. The peptide with a 2-residue spacing (-S(P)-X-X-S-) was by far the best casein kinase I substrate. When the P-Ser residue at Ser7 was replaced with P-Thr, the resulting peptide was still a casein kinase I substrate. However, substitution of Asp or Glu residues at Ser7 led to peptides that were not phosphorylated by casein kinase I. Phosphorylation of one of the other peptides showed that Thr could also be the phosphate acceptor. From these results, we propose that there are substrates for casein kinase I for which prior phosphorylation is a critical determinant of protein kinase action. In these instances, an important recognition motif for casein kinase I appears to be -S(P)/T(P)-Xn-S/T- with n = 2 much more effective than n = 1 or n = 3. Thus, casein kinase I may be involved in hierarchal substrate phosphorylation schemes in which its activity is controlled by the phosphorylation state of its substrates.
Article
Full-text available
X-linked Charcot-Marie-Tooth disease (CMTX) is a form of hereditary neuropathy with demyelination. Recently, this disorder was mapped to chromosome Xq13.1. The gene for the gap junction protein connexin32 is located in the same chromosomal segment, which led to its consideration as a candidate gene for CMTX. With the use of Northern (RNA) blot and immunohistochemistry technique, it was found that connexin32 is normally expressed in myelinated peripheral nerve. Direct sequencing of the connexin32 gene showed seven different mutations in affected persons from eight CMTX families. These findings, a demonstration of inherited defects in a gap junction protein, suggest that connexin32 plays an important role in peripheral nerve.
Article
Full-text available
Intercellular gap junction channels are thought to form when oligomers of connexins from one cell (connexons) register and pair with connexons from a neighboring cell en route to forming tightly packed arrays (plaques). In the current study we used the rat mammary BICR-M1Rk tumor cell line to examine the trafficking, maturation, and kinetics of connexin43 (Cx43). Cx43 was conclusively shown to reside in the Golgi apparatus in addition to sites of cell-cell apposition in these cells and in normal rat kidney cells. Brefeldin A (BFA) blocked Cx43 trafficking to the surface of the mammary cells and also prevented phosphorylation of the 42-kD form of Cx43 to 44- and 46-kD species. However, phosphorylation of Cx43 occurred in the presence of BFA while it was still a resident of the ER or Golgi apparatus yielding a 43-kD form of Cx43. Moreover, the 42- and 43-kD forms of Cx43 trapped in the ER/Golgi compartment were available for gap junction assembly upon the removal of BFA. Mammary cells treated with BFA for 6 h lost preexisting gap junction "plaques," as well as the 44- and 46-kD forms of Cx43 and functional coupling. These events were reversible 1 h after the removal of BFA and not dependent on protein synthesis. In summary, we provide strong evidence that in BICR-M1Rk tumor cells: (a) Cx43 is transiently phosphorylated in the ER/Golgi apparatus, (b) Cx43 trapped in the ER/Golgi compartment is not subject to rapid degradation and is available for the assembly of new gap junction channels upon the removal of BFA, (c) the rapid turnover of gap junction plaques is correlated with the loss of the 44- and 46-kD forms of Cx43.
Article
Full-text available
Severe deafness or hearing impairment is the most prevalent inherited sensory disorder, affecting about 1 in 1,000 children. Most deafness results from peripheral auditory defects that occur as a consequence of either conductive (outer or middle ear) or sensorineuronal (cochlea) abnormalities. Although a number of mutant genes have been identified that are responsible for syndromic (multiple phenotypic disease) deafness such as Waardenburg syndrome and Usher 1B syndrome, little is known about the genetic basis of non-syndromic (single phenotypic disease) deafness. Here we study a pedigree containing cases of autosomal dominant deafness and have identified a mutation in the gene encoding the gap-junction protein connexin 26 (Cx26) that segregates with the profound deafness in the family. Cx26 mutations resulting in premature stop codons were also found in three autosomal recessive non-syndromic sensorineuronal deafness pedigrees, genetically linked to chromosome 13q11-12 (DFNB1), where the Cx26 gene is localized. Immunohistochemical staining of human cochlear cells for Cx26 demonstrated high levels of expression. To our knowledge, this is the first non-syndromic sensorineural autosomal deafness susceptibility gene to be identified, which implicates Cx26 as an important component of the human cochlea.
Article
Full-text available
Immortalized cells from embryonic connexin43 knockout mice (Cx43-/-) and homozygous littermates (Cx43+/+) were cloned and characterized to determine whether the absence of Cx43 function would induce observable phenotypic changes. Cells of the Cx43+/+ clones expressed Cx43 and engaged in gap junctional communication with 10-12 neighboring cells. The Cx43-/- cells were devoid of Cx43 and communicated to less than 1 cell. Electrophysiological analysis indicated that the Cx43-/- cells communicated through Cx45 channels from 8-80-fold less than did the Cx43+/+ subclones, which seemed to communicate through Cx43 and Cx45 channels. The Cx43-/- clones grew at faster rates and to higher saturation densities, had a more spindly morphology, were more refractile, and adhered less well to the substratum than did the Cx43+/+ clones. Reintroducing the Cx43 gene into the Cx43-/- clones resulted in three subclones that communicated to 3-4 cells. Partial restoration of gap junctional communication in the three subclones was accompanied by reduced growth rates and saturation densities (2-fold compared to that of parental Cx43-/- clones) but no reversions in morphology or cell-substratum adhesion. The increased growth rates and saturation densities, altered morphology, and decreased cell adhesion displayed by the Cx43-/- clones reflect a subset of the properties of transformed cells. These studies advance the hypothesis that loss of Cx43 function during development may cause cells to acquire a preneoplastic condition.
Article
Coordinated contractile activation of the heart and resistance to ischemic injury depend, in part, on the intercellular communication mediated by Cx43-composed gap junctions. The function of these junctions is regulated at multiple levels ( assembly to degradation) through phosphorylation at specific sites in the carboxyl terminus (CT) of the Cx43 protein. We show here that the selective permeability of Cx43 junctions is regulated through protein kinase C (PKC)-dependent phosphorylation at serine 368 (S368). Selective permeability was measured in several Cx43-expressing cell lines as the rate constant for intercellular dye diffusion relative to junctional conductance. The selective permeability of Cx43 junctions under control conditions was quite variable, as was the open-state behavior of the comprising channels. Coexpression of the CT of Cx43 as a distinct protein, treatment with a PKC inhibitor, or mutation of S368 to alanine, all reduced (or eliminated) phosphorylation at S368, reduced the incidence of 55- to 70-pS channels, and reduced by 10-fold the selective permeability of the junctions for a small cationic dye. Because PKC activation during preischemic conditioning is cardioprotective during subsequent ischemic episodes, we examined no-flow, ischemic hearts for Cx43 phosphorylated at S368 (pS368). Consistent with early activation of PKC, pS368-Cx43 was increased in ischemic hearts; despite extensive lateralization of total Cx43, pS368-Cx43 remained predominantly at intercalated disks. Our data suggest that the selectivity of gap junction channels at intercalated disks is increased early in ischemia.
Article
Intercellular gap junction channels are thought to form when oligomers of connexins from one cell (connexons) register and pair with connexons from a neighboring cell en route to forming tightly packed arrays (plaques). In the current study we used the rat mammary BICR-M1Rk tumor cell line to examine the trafficking, maturation, and kinetics of connexin43 (Cx43). Cx43 was conclusively shown to reside in the Golgi apparatus in addition to sites of cell-cell apposition in these cells and in normal rat kidney cells. Brefeldin A (BFA) blocked Cx43 trafficking to the surface of the mammary cells and also prevented phosphorylation of the 42-kD form of Cx43 to 44- and 46-kD species. However, phosphorylation of Cx43 occurred in the presence of BFA while it was still a resident of the ER or Golgi apparatus yielding a 43-kD form of Cx43. Moreover, the 42- and 43-kD forms of Cx43 trapped in the ER/Golgi compartment were available for gap junction assembly upon the removal of BFA. Mammary cells treated with BFA for 6 h lost preexisting gap junction "plaques," as well as the 44- and 46-kD forms of Cx43 and functional coupling. These events were reversible 1 h after the removal of BFA and not dependent on protein synthesis. In summary, we provide strong evidence that in BICR-M1Rk tumor cells: (a) Cx43 is transiently phosphorylated in the ER/Golgi apparatus, (b) Cx43 trapped in the ER/Golgi compartment is not subject to rapid degradation and is available for the assembly of new gap junction channels upon the removal of BFA, (c) the rapid turnover of gap junction plaques is correlated with the loss of the 44- and 46-kD forms of Cx43.
Article
Gap junction channels formed by α3 (Cx46) and α8 (Cx50) connexin provide pathways for communication between the fiber cells in the normal transparent lens. To determine the specific role of α3 connexin in vivo, the α3 connexin gene was disrupted in mice. Although the absence of α3 connexin had no obvious influence on the early stages of lens formation and the differentiation of lens fibers, mice homozygous for the disrupted α3 gene developed nuclear cataracts that were associated with the proteolysis of crystallins. This study establishes the importance of gap junctions in maintaining normal lens transparency by providing a cell–cell signaling pathway or structural component for the proper organization of lens membrane and cytoplasmic proteins.
Article
The effects of halothane and ethrane on gap junction-mediated intercellular communication and on membrane excitability were examined in cultured neonatal rat cardiac myocytes using whole-cell voltage-clamp and current-clamp techniques. Excitability was maintained at doses of both anesthetics that reversibly abolished current flow through junctional membranes. The degree of reduction of junctional conductance was a steep function of the dose of anesthetic; complete block occurred at lower aqueous concentrations of halothane than ethrane. The time course for loss of communication was rapid; 90% reduction of initial junctional conductance occurred in less than 15 seconds after exposure to 2 mM halothane or 4 mM ethrane. Recovery of junctional conductance and junctional permeability to intracellularly injected Lucifer yellow was rapid and complete on washout of the anesthetics. As junctional conductance was reduced by halothane or ethrane exposure, unitary conductance of the gap junctional channels remained constant at about 50 pS. Uncoupling by these anesthetics is thus attributable to a decrease in the number of conducting channels rather than to reduction of the channel's unitary conductance. The data are discussed with regard to the possible role of this intercellular communication pathway in the arrhythmias and alterations of conduction velocity and contractility produced by volatile anesthetics.