ArticlePDF Available

Expression and function of Kv1.1 potassium channels in human atria from patients with atrial fibrillation

Authors:
  • Georg-August University Göttingen, Germany

Abstract

Voltage-gated Kv1.1 channels encoded by the Kcna1 gene are traditionally regarded as being neural-specific with no known expression or intrinsic functional role in the heart. However, recent studies in mice reveal low-level Kv1.1 expression in heart and cardiac abnormalities associated with Kv1.1-deficiency suggesting that the channel may have a previously unrecognized cardiac role. Therefore, this study tests the hypothesis that Kv1.1 channels are associated with arrhythmogenesis and contribute to intrinsic cardiac function. In intra-atrial burst pacing experiments, Kcna1-null mice exhibited increased susceptibility to atrial fibrillation (AF). The atria of Kcna1-null mice showed minimal Kv1 family ion channel remodeling and fibrosis as measured by qRT-PCR and Masson's trichrome histology, respectively. Using RT-PCR, immunocytochemistry, and immunoblotting, KCNA1 mRNA and protein were detected in isolated mouse cardiomyocytes and human atria for the first time. Patients with chronic AF (cAF) showed no changes in KCNA1 mRNA levels relative to controls; however, they exhibited increases in atrial Kv1.1 protein levels, not seen in paroxysmal AF patients. Patch-clamp recordings of isolated human atrial myocytes revealed significant dendrotoxin-K (DTX-K)-sensitive outward current components that were significantly increased in cAF patients, reflecting a contribution by Kv1.1 channels. The concomitant increases in Kv1.1 protein and DTX-K-sensitive currents in atria of cAF patients suggest that the channel contributes to the pathological mechanisms of persistent AF. These findings provide evidence of an intrinsic cardiac role of Kv1.1 channels and indicate that they may contribute to atrial repolarization and AF susceptibility.
ORIGINAL CONTRIBUTION
Expression and function of Kv1.1 potassium channels in human
atria from patients with atrial fibrillation
Edward Glasscock
1
Niels Voigt
2,3
Mark D. McCauley
4
Qiang Sun
2,3
Na Li
4
David Y. Chiang
4
Xiao-Bo Zhou
3
Cristina E. Molina
2
Dierk Thomas
3
Constanze Schmidt
3
Darlene G. Skapura
4
Jeffrey L. Noebels
5,6,7
Dobromir Dobrev
2,3
Xander H. T. Wehrens
4
Received: 19 December 2014 / Revised: 2 July 2015 / Accepted: 3 July 2015 / Published online: 11 July 2015
ÓSpringer-Verlag Berlin Heidelberg 2015
Abstract Voltage-gated Kv1.1 channels encoded by the
Kcna1 gene are traditionally regarded as being neural-
specific with no known expression or intrinsic functional
role in the heart. However, recent studies in mice reveal
low-level Kv1.1 expression in heart and cardiac abnor-
malities associated with Kv1.1-deficiency suggesting that
the channel may have a previously unrecognized cardiac
role. Therefore, this study tests the hypothesis that Kv1.1
channels are associated with arrhythmogenesis and con-
tribute to intrinsic cardiac function. In intra-atrial burst
pacing experiments, Kcna1-null mice exhibited increased
susceptibility to atrial fibrillation (AF). The atria of Kcna1-
null mice showed minimal Kv1 family ion channel
remodeling and fibrosis as measured by qRT-PCR and
Masson’s trichrome histology, respectively. Using RT-
PCR, immunocytochemistry, and immunoblotting, KCNA1
mRNA and protein were detected in isolated mouse car-
diomyocytes and human atria for the first time. Patients
with chronic AF (cAF) showed no changes in KCNA1
mRNA levels relative to controls; however, they exhibited
increases in atrial Kv1.1 protein levels, not seen in
paroxysmal AF patients. Patch-clamp recordings of iso-
lated human atrial myocytes revealed significant dendro-
toxin-K (DTX-K)-sensitive outward current components
that were significantly increased in cAF patients, reflecting
a contribution by Kv1.1 channels. The concomitant
increases in Kv1.1 protein and DTX-K-sensitive currents in
atria of cAF patients suggest that the channel contributes to
the pathological mechanisms of persistent AF. These
findings provide evidence of an intrinsic cardiac role of
Kv1.1 channels and indicate that they may contribute to
atrial repolarization and AF susceptibility.
Keywords Voltage-gated potassium channels Atrial
fibrillation Dendrotoxin-K Kcna1 Kv1.1
Introduction
Atrial fibrillation (AF) is the most common sustained car-
diac arrhythmia and associated with significant cardiovas-
cular morbidity and mortality, but its underlying molecular
basis remains only partially understood [47]. During AF,
atrial cells fire rapidly at rates up to tenfold faster than
normal, producing uncoordinated atrial activity and irreg-
ular ventricular contraction, which can lead to blood clot
formation and stroke [21]. Alteration of ion channel
Electronic supplementary material The online version of this
article (doi:10.1007/s00395-015-0505-6) contains supplementary
material, which is available to authorized users.
&Edward Glasscock
aglas1@lsuhsc.edu
1
Department of Cellular Biology and Anatomy, Louisiana
State University Health Sciences Center, 1501 Kings
Highway, P.O. Box 33932, Shreveport, LA 71130-393, USA
2
Faculty of Medicine, Institute of Pharmacology, University
Duisburg-Essen, Essen, Germany
3
Division of Experimental Cardiology, Medical Faculty
Mannheim, University of Heidelberg, Mannheim, Germany
4
Department of Molecular Physiology and Biophysics, and
Medicine/Cardiology, Cardiovascular Research Institute,
Baylor College of Medicine, Houston, TX, USA
5
Departments of Neurology, Baylor College of Medicine,
Houston, TX, USA
6
Departments of Neuroscience, Baylor College of Medicine,
Houston, TX, USA
7
Departments of Molecular and Human Genetics, Baylor
College of Medicine, Houston, TX, USA
123
Basic Res Cardiol (2015) 110:47
DOI 10.1007/s00395-015-0505-6
function by atrial remodeling or genetic mutation can
provide a pro-fibrillatory electrophysiological substrate
conducive to AF [13,22]. A variety of conditions can cause
ion channel remodeling predisposing to AF, including
congestive heart failure and acute myocardial infarction
[23]. In addition, AF itself can cause electrical remodeling
that promotes persistent fibrillation and thereby auto-per-
petuates the arrhythmia. The underlying mechanisms for
this transition likely involve altered channel expression and
function [22,23,50]. Although AF is primarily a sporadic
condition, population-based studies and rare familial kin-
dreds have shown that it has a significant genetic compo-
nent [19,35]. In families with monogenic AF subtypes, the
majority of genes implicated encode subunits of voltage-
gated potassium and sodium channels [19].
Here Kv1.1 voltage-gated potassium channels were
investigated for a role in AF. Kv1.1 channels, encoded by
the Kcna1 gene, exhibit widespread expression throughout
the brain and peripheral nervous system and their dys-
function leads to neurological diseases including epilepsy
and episodic ataxia type 1 [29]. Kv1.1 channels have tra-
ditionally been regarded as predominantly neural-specific
with no known expression or function in the heart. How-
ever, mice lacking Kv1.1 channels exhibit atrioventricular
cardiac conduction abnormalities and bradyarrhythmia
phenotypes that appear to emanate from seizures and
abnormal vagal activity [10,11]. In addition, these previ-
ous studies detected low levels of Kcna1 mRNA and pro-
tein in mouse heart suggesting that Kv1.1 channels may
also contribute to the intrinsic function of the heart [11]. If
so, alteration of Kv1.1 channel function could lead to
independent dual arrhythmia phenotypes in brain and heart.
In this work, a combination of electrophysiological
techniques and molecular analyses was used to evaluate the
contribution of Kv1.1 channels to basal cardiac function
and potential arrhythmia development. Two main
hypotheses were tested: (1) that Kv1.1 channel perturbation
in mice causes arrhythmia susceptibility, and (2) that
dysregulation of Kv1.1 channels in the human heart may be
important for arrhythmogenesis. Our experiments show
that the absence of Kv1.1 channels predisposes the mouse
heart to AF without drastic remodeling of related K?
channel subunits and fibrotic structural changes. Expres-
sion analyses in isolated mouse myocytes demonstrate the
presence of Kv1.1 mRNA and protein in heart apart from
neural tissue. Molecular analyses detect the first clear
evidence of Kv1.1 expression in human atria, and show
that Kv1.1 channels exhibit expression changes in patients
with chronic AF suggestive of pathophysiological channel
remodeling. In addition, patch-clamp recordings of isolated
human atrial myocytes reveal significant DTX-K-sensitive
components that are doubled in patients with cAF,
indicative of a contribution by Kv1.1 channels. Taken
together this work finds a previously unrecognized cardiac
role for the Kcna1 gene and Kv1.1 channels in regulating
atrial repolarization and arrhythmia susceptibility.
Methods
Animals and genotyping
Kcna1
-/-
mice carry null alleles of the Kcna1 gene
resulting from targeted deletion of the open reading frame,
as described [36]. The mice are maintained on a Tac:N:-
NIHS-BC background. Animals were housed at 22 °C, fed
ad libitum, and submitted to a 12 h light/dark cycle. For
surgeries, mice were anaesthetized using 1.5–2 % isoflu-
rane in 95 % O
2
. Animals were euthanized for expression
and tissue analysis using inhaled isoflurane overdose. All
procedures were performed in accordance with the guide-
lines of the National Institutes of Health, as approved by
the Animal Care and Use Committee of Baylor College of
Medicine.
Genomic DNA was isolated by enzymatic digestion of
tail clips using Direct-PCR Lysis Reagent (Viagen Biotech,
Los Angeles, CA, USA). The genotypes of Kcna1 mice
were determined by performing PCR amplification of
genomic DNA using three allele-specific primers: a
mutant-specific primer (50-CCTTCTATCGCCTTCTT
GACG-30), a wild-type-specific primer (50-GCCTCTGA
CAGTGACCTCAGC-30), and a common primer (50-GC
TTCAGGTTCGCCACTCCCC-30). The PCR yielded
amplicons of *337 bp for the wild-type allele and
*475 bp for the null allele.
Intracardiac electrophysiology in mice
In vivo electrophysiology studies were performed in
knockout and wild-type mice of both sexes, as per prior
established protocols [15,37]. Atrial and ventricular
intracardiac electrograms were recorded simultaneously
using a 1.1F octapolar catheter (EPR-800, Millar Instru-
ments, Houston, TX, USA) inserted via the right jugular
vein. Surface ECG and intracardiac electrophysiology
parameters were assessed at baseline. Right atrial pacing
was performed using 2-ms current pulses at 800 lA
delivered by an external stimulator (STG-3008, Multi
Channel Systems, Reutlingen, Germany). AF inducibility
was determined using an overdriving pacing protocol, and
AF was defined as the occurrence of rapid and fragmented
atrial electrograms with irregular AV-nodal conduction and
ventricular rhythm for at least 1 s. To be counted as AF
positive, a mouse had to exhibit AF in response to at least
two out of three pacing trials. For Kcna1-null mice, the
mean AF duration was determined by calculating the
47 Page 2 of 15 Basic Res Cardiol (2015) 110:47
123
average elapsed time of all observed AF episodes. The
stimulation protocols used for intracardiac burst pacing are
summarized in Supplemental Table S1.
Mouse RNA extraction, cDNA synthesis, and real-
time PCR
Similarly aged Kv1.1-knockout mice (n=5;
112 ±5 days old) and wild-type control mice (n=5;
121 ±5 days old) were euthanized using isoflurane
inhalation and their left atria quickly excised. The tissue
was immediately placed in ice-cold TRIzol reagent,
homogenized, and the total RNA extracted according to the
manufacturer’s protocol (Invitrogen, Carlsbad, CA, USA).
Following resuspension in water, RNA samples were
DNase treated using the DNA-free Kit (Applied Biosys-
tems, Carlsbad, CA, USA). The quantity of total RNA was
measured and the quality checked by agarose gel
electrophoresis.
First-strand cDNA was synthesized from 250 ng of total
RNA using the Phusion RT-PCR Kit with oligo(dT) primers
(ThermoScientific, Waltham, MA, USA). Before proceeding to
real-time PCR, the quality of cDNA and the tissue genotype
was verified by 40 cycles of PCR amplification using primers
specific for the GAPDH and Kcna1 genes, followed by agarose
gel electrophoresis. A 982-bp GAPDH-specific band was
amplified using the following primers: forward, 50-
TGAAGGTCGGTGTGAACGGATTTGGC-30; reverse, 50-
ATGTAGGCCATGAGGTCCACCAC-30. A 710-bp Kcna1-
specific band was amplified using the following primers: for-
ward, 50-GCATCGACAACACCACAGTC-3; reverse, 50-
CGGCGGCTGAGGTCACTGTCAGAGGCTAAGT-30.The
lack of GAPDH PCR products in –RT controls confirmed the
absence of genomic DNA contamination, while the presence or
absence of Kcna1 amplicons confirmed the tissue genotype and
verified the absence of Kcna1 mRNA in knockouts.
To quantify the relative gene expression patterns in left
atria of knockout and control animals, real-time RT-PCR of
first strand cDNA was performed with TaqMan gene
expression assays using the 7500 Real-Time PCR System
(Applied Biosystems). TaqMan gene expression assays
were designed and preoptimized by Applied Biosystems
for the detection of Kcna1 (Mm00439977_s1), Kcna2
(Mm00434584_s1), Kcna3 (Mm00434599_s1), Kcna4
(Mm00445241_s1), Kcna5 (Mm00524346_s1), Kcna6
(Mm00496625_s1), Kcna7 (Mm01197268_m1), Kcnab1
(Mm00440017_m1), Kcnab2 (Mm01260263_m1), Kcnab3
(Mm01337146_m1), col6a1 (Mm00487160_m1), and
Hprt1 (Mm00446968_m1). Each assay consisted of an
unlabeled gene-specific PCR primer pair and a TaqMan
probe with a fluorescent FAM dye label and a minor
groove binder moiety on the 50end and a nonfluorescent
quencher on the 30end. Individual PCRs were performed in
triplicate using 100 ng of cDNA and TaqMan Gene
Expression Master Mix (Applied Biosystems). No template
and –RT reactions were included as negative controls to
verify the absence of contamination leading to unwanted
PCR amplification and detection. The reactions were
cycled at 50 °C for 2 min, 95 °C for 10 min, and then 40
cycles of 95 °C for 15 s and 60 °C for 1 min. Spectral data
were collected and analyzed with SDS 1.3 software (Ap-
plied Biosystems) using a manual threshold of 0.1, auto-
matic baseline detection, and automatic outlier removal.
The data were analyzed using the threshold cycle (C
t
)
relative quantification method [16]. Hprt1 was used as a
reference gene for normalization since it showed stable and
reliable expression in our samples. GAPDH was also
evaluated as a reference gene but its expression was more
variable than Hprt1, so it was excluded from the analysis.
The C
t
values were averaged and then used to calculate the
expression levels relative to Hprt1 using the formula 2DCt
X 100, where 2DCtcorresponds to the difference in the
threshold cycle of the gene of interest versus Hprt1. In two
instances, once for Kcna6 and once for Kcnab3 (both in wild-
type samples), the assays failed to amplify across the entire
triplicate, even when the experiment was repeated, likely due
to extremely low levels of gene expression. In both cases, a
value of zero was imputed for the 2DCtmeasurement for the
given samples. In rare instances where triplicates produced
only one successful replicate, the C
t
value used for data
analysis was derived from the single working reaction,
assuming that the measurement was similar to the other
biological replicates. Values for Kcna1 levels in knockout
tissue were imputed as zero since the TaqMan assay used for
detection is uninformative in knockouts; the Kcna1 assay is
targeted to the 50-UTR of the gene which is still present in
knockouts despite the absence of the open reading frame. As
stated above, all knockout tissues were verified to be nega-
tive for Kcna1 mRNA using 40 cycles of RT-PCR followed
by agarose gel electrophoresis.
Mouse histology
Whole hearts from 4- to 6-month-old mice were excised,
submerged in 1 M KCl for 45–60 s, rinsed briefly in PBS,
and then fixed in 10 % neutral buffered formalin for 24 h.
Following fixation, hearts were embedded in paraffin,
longitudinally sectioned (5 lm), and stained with Masson’s
trichrome for fibrosis. The percentage of fibrotic atrial
tissue was quantified with Adobe Photoshop using previ-
ously described methods [8]. In brief, images were adjusted
using the selective color command to intensify the contrast
between reds and blues. The color range function was then
used to generate histograms which counted the pixel area
for each of the color components. The fraction of tissue
fibrosis was then calculated by dividing the sum of the cyan
Basic Res Cardiol (2015) 110:47 Page 3 of 15 47
123
and blue pixel areas by the total pixel area. For each ani-
mal, percentage fibrosis was estimated by averaging mea-
surements from left and right atria in similarly oriented
single longitudinal sections. Fibrosis percentages appeared
similar between left and right atria for each genotype. For
one knockout and one control animal, fibrosis was mea-
sured in only the left or right atria due to the loss of tissue
during processing.
Cardiomyocyte isolation and expression analyses
C57BL/6 mice (3 months old) were used for cell isolation
as described previously [15]. In brief, following isoflurane
anesthesia, the heart was removed from mice. After dis-
secting, the heart was cannulated through the aorta and
perfused on a Langendorff apparatus with 0 Ca
2?
Tyrode,
then 0 Ca
2?
Tyrode containing 0.02 mg/mL Liberase TH
Research Grade (Roche Applied Science, Penzberg, Ger-
many) for 10–15 min at 37 °C. After digestion, atria and
ventricle were separated and placed in KB solution
(90 mmol/L KCl, 30 mmol/L K
2
HPO
4
, 5 mmol/L MgSO
4
,
5 mmol/L pyruvic acid, 5 mmol/L b-hydroxybutyric acid,
5 mmol/L creatine, 20 mmol/L taurine, 10 mmol/L glu-
cose, 0.5 mmol/L EGTA, 5 mmol/L HEPES; pH 7.2), then
minced thoroughly and agitated gently, and filtered through
a 210 lm mesh. The isolated myocytes were spun down at
800 rpm for 5 min. After removing supernatant, the cells
were used immediately for expression analysis or stored at
-80 °C for biochemical studies later.
Total RNA was isolated using Direct-zolTM RNA
MiniPrep (Zymo Research, Irvine, CA, USA) according to
the manufacturer’s instructions. In column DNase I digestion
was performed to remove DNA contamination. First-stand
cDNA was synthesized from 300 ng of total RNA using
iScript
TM
cDNA Synthesis Kit (Bio-Rad, Hercules, CA,
USA). PCR was then performed as described above in
‘‘ Mouse RNA extraction, cDNA synthesis, and real-time
PCR using the same primers and amplification protocol.
Quantitative real-time PCR (qRT-PCR) was performed as
previously reported with modifications [28]. Briefly, qRT-
PCR was performed in triplicates with PerfeCTa
Ò
SYBR
Ò
Green FastMix (Quanta BioSciences, Inc., Gaithersburg,
MD, USA) in 96-well plates using Mastercycler ep realplex
(Eppendorf, Hamburg, Germany). The following program
was run to amplify the products: (1) 95 °C for 5 min, (2) 45
cycles of 95 °C for 5 s, 60 °C for 10 s, 72 °C for 1 s, (3)
melting curve ramp from 65 °Cto95 °C at 0.1 °C per sec-
ond. mRNA expression levels were compared using the
relative CT (cycle number) method after normalization to
Rpl7. Primer sequences for Kcna1 were the following: for-
ward, 50-GAGAATGCGGACGAGGCTTC-30and reverse,
50-CCGGAGATGTTGATTACTACGC-30. For Rpl7, the
primers had the following sequence: forward, 50GAAGCT
CATCTATGAGAAGGC-30and reverse, 50-AAGACGAA
GGAGCTGCAGAAC-30.
For immunocytochemistry, isolated myocytes were
added to cover glasses coated with 20 lg/ml laminin in
PBS for 30 min before fixation with 4 % formalin for
15 min. After washing with PBS 3 times, the myocytes
were permeabilized with 0.1 % Triton X in PBS for
10 min, washed another 3 times with PBS, and blocked
with 1 % normal goat serum (NGS) in PBS for 1 h. After
blocking, the samples were incubated overnight at 4 °C
with antibodies against Kv1.1 (K20/78, NeuroMab, Davis,
CA, USA; 1:100) and JPH2 (custom-made [26]; 1:100)
diluted in 1 % NGS and 1 % bovine serum albumin (BSA)
in PBS. Next day, the cover glasses were washed with PBS
3 times and incubated with Alexa Fluor
Ò
568 Goat Anti-
Mouse IgG and Alexa Fluor
Ò
488 Goat Anti-Rabbit IgG
(#A11004, Invitrogen, Carlsbad, CA, USA; 1:2000 for
both) in 1 % NGS/BSA in PBS for 1 h at room tempera-
ture. Afterward, the cover glasses were washed with PBS 3
times and mounted on glass slide with Vectashield media
with DAPI (#H-1200, Vector Laboratories, Burlingame,
CA, USA). Fluorescence images were taken with the Zeiss
LSM510 confocal microscope (Jena, Germany).
Human tissue samples
Right atrial appendages were obtained from 32 patients in
sinus rhythm (SR), 14 in paroxysmal AF (pAF) and 23 in
chronic AF (cAF), undergoing open-heart surgery
(Table 1). Experimental protocols were approved by the
ethics committee of the Medical Faculty Mannheim,
University of Heidelberg (#2011-216 N-MA) and per-
formed in accordance with the Declaration of Helsinki.
Each patient gave written informed consent. After excision,
atrial appendages were used for either myocyte isolation (6
SR, 6 cAF) or snap-frozen in liquid-nitrogen for bio-
chemical studies (26 SR, 14 pAF, 17 cAF patients).
Human RNA and protein biochemistry
RNA isolation, reverse-transcription and real-time PCR
were performed as described [43]. For initial detection of
KCNA1 mRNA, cDNA was PCR-amplified using 35 cycles
and primers specific for the KCNA1 gene, followed by
agarose gel electrophoresis. A 232-bp KCNA1-specific
band was amplified using the following primers: forward,
50-CATCGTGGAAACGCTGTGTAT-3; reverse, 50AAC
CCTTACCAAGCGGATGAC-30. Commercial primers
were used for real-time PCR detection of human KCNA1
mRNA expression in patients (Hs00264798_s1, Life
Technologies, Foster City, CA, USA). KCNA1 mRNA
expression was normalized to HPRT1 (Hs01003267_m1;
Life Technologies).
47 Page 4 of 15 Basic Res Cardiol (2015) 110:47
123
Protein levels of Kv1.1 (1:1000; rabbit polyclonal anti-
Kv1.1; ARP34923_P050; Aviva Systems Biology, San
Diego, CA, USA) were quantified by Western blotting and
normalized to GAPDH (1:200,000; HyTest, Turku, Fin-
land) as described [43]. Peroxidase-conjugated goat anti-
rabbit (1:5000; Sigma-Aldrich, St. Louis, MO, USA) and
goat anti-mouse (1:50,000; Sigma-Aldrich) were used as
secondary antibodies and visualized by chemifluorescense
(GE Healthcare, Chalifont St. Giles, UK). AIDA Image
Analyzer Software (raytest, Straubenhardt, Germany) was
used for analysis.
For immunocytochemistry, cells (from 3 patients in SR;
8 cells/patient) were fixed with paraformaldehyde (PFA;
2 %) for 15 min. After centrifugation at 750 rpm for
5 min, PFA was removed, the myocytes were washed, and
PFA was neutralized with Glycin (0.1 M). Cells were then
washed 3 times with phosphate-buffered saline (PBS) and
permeabilized for 5 min with Triton X-100 (0.5 %) diluted
in PBS. Cells were rinsed 3 times with PBS and then
blocked with 1 % BSA in PBS. Myocytes were then
labeled with the primary antibody (rabbit polyclonal anti-
Kv1.1 (1:1000); ARP34923_P050; Aviva Systems
Table 1 Patient characteristics SR PAF CAF
Patients, n32 14 23
Gender, m/f 24/8 10/4 15/8
Age, years 64.09 ±11.2 66.9 ±10.1 67.78 ±9.41
Body mass index, kg/m
2
28.09 ±5.79 29.54 ±7.53 29.23 ±4.21
CAD, n(%) 17 (53) 4 (29) 8 (35)
AVD, n(%) 8 (25) 6 (43) 6 (26)
MVD, n(%) 0 (0) 1 (7) 5 (22)
CAD ?AVD, n(%) 7 (22) 4 (29) 5 (22)
Hypertension, n(%) 28 (88) 14 (100) 22 (96)
Diabetes, n(%) 7 (22) 4 (29) 8 (35)
Hyperlipidemia, n(%) 29 (91) 12 (86) 17 (74)
LVEF
Normal, n(%) 16 (50) 6 (43) 11 (48)
Mildly reduced, n(%) 10 (31) 2 (14) 3 (13)
Moderately reduced, n(%) 4 (13) 4 (29) 6 (26)
Severely reduced, n(%) 1 (3) 2 (14) 3 (13)
b-Blockers, n(%) 20 (62) 14 (100) 19 (83)
Digitalis, n(%) 2 (6) 2 (14) 7 (30)
Amiodarone, n(%)
#
0 (0) 0 (0) 1 (7)
Other AADs, n(%) 0 (0) 0 (0) 0 (0)
ACE inhibitors, n(%) 19 (59) 8 (57) 12 (52)
AT1 blockers, n(%) 6 (19) 2 (14) 3 (13)
Dihydropyridines, n(%) 8 (25) 4 (29) 3 (13)
Diuretics, n(%) 9 (28) 10 (71) 11 (48)
Nitrates, n(%) 1 (3) 2 (14) 3 (13)
Lipid-lowering drugs, n(%) 22 (69) 11 (79) 14 (61)
Biguanides, n(%)
#
2 (7) 2 (18) 1 (7)
Sulfonylurea derivatives, n(%)
#
1 (4) 0 (0) 0 (0)
Insulin, n(%)
#
0 (0) 3 (27) 2 (14)
OAC, n(%)
#
0 (0) 5 (45)** 13 (93)***
Antiplatelet drugs, n(%)
#
23 (88) 8 (73) 6 (43)*
AAD antiarrhythmic drug, ACE angiotensin converting enzyme, AT angiotensin receptor, AVD aortic valve
disease, CAD coronary artery disease, CAF chronic atrial fibrillation, LVEF left ventricular ejection fraction
(normal, C55 %; mild impairment, 45–54 %; moderate impairment, 30–44 %; severe impairment,\30 %),
MVD mitral valve disease, OAC oral anticoagulation, PAF paroxysmal atrial fibrillation, SR sinus rhythm
*P\0.05, ** P\0.01, *** P\0.001 versus SR from Fisher exact test followed by Bonferroni multiple
comparisons procedure for categorical variables
#
Data were not available for 6 SR, 3 pAF and 9 cAF patients
Basic Res Cardiol (2015) 110:47 Page 5 of 15 47
123
Biology, San Diego, CA, USA), which was diluted in PBS
containing 1 % BSA. After an overnight incubation at
4°C, the myocytes were washed 3 times using 1 % BSA in
PBS and then incubated with Fluorescein (FITC, excitation
488 nm, emission 520 nm) anti-rabbit IgG to reveal the
Kv1.1 staining. After 3 successive 5-min washes using 1 %
BSA in PBS, the cells were mounted with Mowiol med-
ium. Images were acquired using a CAIRN Spinning Disk
confocal microscope.
Patch-clamp experiments
Atrial myocytes were isolated using a standard protocol
and were suspended in storage solution (mmol/L: KCl 20,
KH
2
PO4 10, glucose 10, K-glutamate 70, b-hydroxybu-
tyrate 10, taurine 10, EGTA 10, albumin 1, pH =7.4) [44].
Membrane currents were measured in whole-cell ruptured-
patch configuration using voltage clamp. pClamp-Software
V10.2 (Molecular Devices, Sunnyvale, USA) was used for
data acquisition and analysis. Borosilicate glass micro-
electrodes had tip resistances of 2–5 MXwhen filled with
pipette solution (mmol/L: EGTA 0.02, GTP-Tris 0.1,
HEPES 10, K-aspartate 92, KCl 48, Mg-ATP 1, Na
2
-ATP
4; pH =7.2). Seal resistances were 4–8 GX. Series resis-
tance and cell capacitance were compensated. The series
resistance was kept below 10 MXand was compensated by
at least 50 %. With a current amplitude of 1 nA, the
potential for error amounted to \5 mV. To control for
myocyte-size variability, currents are expressed as densi-
ties (pA/pF). Myocytes were superfused at 37 °Cwitha
bath solution containing (mmol/L): CaCl
2
2, glucose 10,
HEPES 10, KCl 4, MgCl
2
1, NaCl 140, probenecid 2;
pH =7.4. I
Ca,L
was blocked by adding CdCl
2
(0.3 mmol/
L) to the bath solution. Drugs were applied via a rapid-
solution exchange system (ALA Scientific Instruments,
Farmingdale, USA). Dendrotoxin-K (10 nmol/L; Alomone
Labs, Jerusalem, Israel) was used to block Kv1.1 currents
[31].
Statistical analysis
Differences between group means for continuous data were
compared by unpaired two-tailed Student’s ttest. Differ-
ences between mean mRNA levels in mouse ventricular
and atrial myocytes were compared using a paired ttest.
Categorical data were analyzed with Fisher’s exact test.
Data are mean ±SEM. P\0.05 was considered statisti-
cally significant. In statistical comparisons of myocyte
electrophysiology data, patients may contribute more than
one observation to each sub-analysis suggesting that
observations are not necessarily independent. Within-pa-
tient correlations were not taken into account in statistical
comparisons due to the low sample size.
Results
Increased susceptibility to pacing-induced AF
in Kcna1-null mice
In our previous studies using video electroencephalography
(EEG) combined with ECG, unequivocal spontaneous AF
was not identified in the ambulatory ECGs of Kcna1-null
mice [11]. To test for AF inducibility, Kcna1-null mice
(age 4.0 ±0.3 months) and WT controls (age
4.1 ±0.3 months) were subjected to intracardiac right
atrial burst pacing stimulation. Simultaneous surface elec-
trocardiograms (ECG) and intracardiac atrial and ventric-
ular electrograms were recorded to monitor the occurrence
of atrial arrhythmias (Fig. 1a). AF was characterized by the
combination of a lack of P waves combined with the
presence of irregular RR intervals in lead I of the surface
ECG (Fig. 1b). Burst pacing induced AF more frequently
in Kcna1-null mice (40 %, 5 of 12) than WT mice (0 %, 0
of 10; P\0.05; Fig. 1c). The average duration of AF
episodes in Kcna1-null mice was 8.0 ±2.9 s, and normal
sinus rhythm always resumed spontaneously. Experiments
using ventricular burst pacing protocols did not show
obvious differences in ventricular arrhythmia susceptibility
between genotypes.
Baseline recordings during anesthesia did not reveal any
significant differences in surface ECG characteristics
between Kcna1-null and WT animals, except for slight but
significant shortening of the QRS interval in null mice
(6.9 ±0.2 ms in Kcna1-null mice versus 7.8 ±0.4 ms in
WT; P\0.05; Table 2). Cardiac conduction system
properties also appeared unaltered, since no significant
differences were found in the sinus node recovery time,
right atrial effective refractory period, and atrioventricular
node effective refractory period (Table 2). The lack of
obvious baseline changes in atrioventricular node function
and sinus cycle length in Kcna1-null mice suggests a
possible neural origin for the increased frequency of AV
conduction blocks found previously in this model; how-
ever, autonomic inputs remain intact in our preparations
and could mask latent cardiac-intrinsic differences. Heart
mass was compared between genotypes to rule out any
effects due to cardiac hypertrophy or remodeling. Hearts of
Kcna1-null mice tended to be larger as measured by
absolute heart mass and heart mass-to-body mass ratio, but
this trend did not reach significance (Table 2).
Minimal Kv1 channel and fibrotic structural
remodeling in atria of Kcna1-null mice
Next, mRNA expression analysis was used to determine
whether the increased AF susceptibility in Kcna1-null mice
47 Page 6 of 15 Basic Res Cardiol (2015) 110:47
123
was potentially caused by compensatory expression chan-
ges in related Kv1.x potassium channel a-subunits, such as
Kcna5 which has been linked to AF [25,52]. Real-time
PCR was performed on atrial tissue from Kcna1-null and
WT mice to compare mRNA expression levels for the
genes Kcna1-7and Kcnab1-3, which encode the Kv1.1-
Kv1.7 pore-forming a-subunits and their associated
Kvbeta1-3 accessory b-subunits, respectively. In WT mice,
measurable expression levels were detected for all seven a-
subunits and all three b-subunits. Among the a-subunits,
Kcna5 was expressed at the highest level followed by
Kcna4 and Kcna7, while Kcna1,Kcna2,Kcna3, and Kcna6
were expressed at similarly low levels (Kcna5 Kc-
na4 [Kcna7 [Kcna1 &Kcna2 &Kcna3 &Kcna6).
There were no substantial compensatory changes in gene
expression levels for the Kv1.2–Kv1.7 a-subunits in Kc-
na1-null mice (age 3.7 ±0.2 months; Fig. 2a). Among the
b-subunits, all were expressed at similarly low levels, and
the Kcnab1 subunit showed a modest but significant 31 %
decrease (P\0.05). However, the overall lack of major
Kv1.x channel subunit remodeling suggests that the
observed increase in AF susceptibility is likely directly
related to Kv1.1 deficiency and not attributable to com-
pensatory dysregulation of related Kv1 subunits.
Since fibrotic structural remodeling can provide a sub-
strate conducive to AF, the hearts of Kcna1-null mice were
examined for evidence of increased fibrillar collagen
deposits. First, transcript levels of the collagen, type VI,
alpha 1 (col6a1) gene were measured as an indicator of
fibrosis. Col6a1 expression levels were significantly
increased by more than 50 % in Kcna1-null atria relative to
WT controls (Fig. 2b; P\0.05). To determine whether
this increase in collagen mRNA correlated with an increase
WT
e
cn
edi
c
n
i
FA
10 12
*
100%
80%
60%
40%
20%
0%
KO
WT
Kcna1-null
(AF onset)
Kcna1-null
(AF termination)
Surface
Atrial
Ventricular
A
BC
77 87 99 100 100 95 109 98 88 93 80 95 92
200 ms
Fig. 1 Mice lacking Kv1.1
channels are vulnerable to
pacing-induced AF.
aRepresentative simultaneous
recordings of surface ECG (lead
I) and intracardiac atrial and
ventricular electrograms after
burst pacing in wild type (WT)
and Kcna1-null (KO) mice. In
the KO animal shown, pacing
produced AF (AF onset) that
lasted about 13 s before
returning to normal sinus
rhythm (AF termination).
bDuring inducible AF in KO
animals, the surface ECG
exhibited absent P waves and
irregular RR intervals, which
are shown labeled in
milliseconds. cKO mice
showed significantly higher
incidence of pacing-induced AF
than WT controls. *P\0.05
Table 2 Baseline ECG parameters in WT and Kcna1-null mice
WT (n=10) KO (n=12) P
Age, months 4.0 ±0.3 4.1 ±0.3 0.68
Heart mass, g
a
160 ±11 173 ±12 0.48
Heart:body ratio, mg/g
a
4.8 ±0.2 5.3 ±0.3 0.23
PQ, ms 27.0 ±0.6 27.0 ±1.2 1.00
QRS, ms 7.8 ±0.4 6.9 ±0.2 0.04
QT, ms 34.0 ±1.6 34.8 ±1.3 0.69
QTc, ms 57.2 ±1.6 57.6 ±1.7 0.88
RR, ms 96.8 ±2.1 98.3 ±2.6 0.67
SCL, ms 96.7 ±2.0 98.3 ±2.5 0.65
AV, ms 33.4 ±0.9 33.8 ±0.9 0.73
SNRT, ms 122.0 ±6.2 125.5 ±8.4 0.75
RA ERP, ms 28.7 ±2.0 31.7 ±2.1 0.34
AV ERP, ms 40.1 ±1.9 41.5 ±3.2 0.75
Data are expressed as mean ±SEM. Student’s ttest used to compare
intragroup differences. Pvalues between WT and KO mice are shown
SCL sinus cycle length, AV atria-to-ventricle conduction time, SNRT
sinus node recovery time, RA ERP right atrial effective refractory
period, AV ERP atrioventricular node effective refractory period
a
Data derived from 9 KO and 6 WT animals
Basic Res Cardiol (2015) 110:47 Page 7 of 15 47
123
in fibrosis, Masson’s trichrome staining of atria was per-
formed to label collagen fibers (Fig. 2c). Quantification of
the percentage of atrial fibrotic tissue showed that Kcna1-
null hearts (1.9 ±0.7 %) tended to have about twice as
much collagen as age-matched wild-type controls
(1.0 ±0.6 %), but the results were highly variable
between animals and did not reach statistical significance
(Fig. 2d; P=0.35). Thus, atrial arrhythmogenesis in Kc-
na1-null hearts is not clearly correlated with increased
atrial fibrosis.
Kv1.1 channels are expressed in isolated mouse
cardiomyoctes
To verify that Kcna1 is expressed in cardiomyocytes and
not only intracardiac neurons, individual atrial and ven-
tricular myocytes were isolated from mouse heart and
analyzed for Kv1.1 mRNA and protein. RT-PCR revealed
the unequivocal presence of Kv1.1 mRNA in isolated atrial
myocytes, but ventricular transcripts were below the limit
of detection (Fig. 3a). Using the more sensitive technique
of real-time PCR to quantify relative mRNA levels, Kv1.1
transcripts were detected in both atrial and ventricular
cells, but the abundance in atrial myocytes was about ten-
fold higher (Fig. 3b; P=0.07). To determine whether
Kcna1 expression could be detected at the protein level in
individual mouse myocytes, immunocytochemistry was
performed. Kv1.1 immunoreactivity was observed in both
atrial and ventricular myocytes (Fig. 3c, d); however, the
Kv1.1-positive ventricular staining appeared much less
intense overall. Importantly, Kv1.1 immunoreactivity was
absent in cardiomyocytes from Kcna1-null mice demon-
strating the specificity of the antibody labeling (Fig. 3c, d).
Kv1.1 channels are expressed in human heart
and upregulated in chronic AF
The increased AF susceptibility in the Kcna1-null mouse
model suggests that Kv1.1 channels may play a role in AF
pathophysiology in patients. However, the expression of
KCNA1 has never been demonstrated in human heart nor
associated with arrhythmia in humans. Using RT-PCR,
KCNA1 mRNA was detected in human atrial samples
confirming its presence in human cardiac tissue for the first
time (Fig. 4a). Since KCNA1 transcripts were measurable
in human atria, mRNA levels of KCNA1 were quantified in
patients with paroxysmal (pAF) and chronic AF (cAF) to
look for disease-associated alterations. Using real-time RT-
PCR, KCNA1 mRNA expression levels were found to be
significantly increased in pAF patients but not in cAF
individuals relative to controls in sinus rhythm (SR)
(Fig. 4b; P\0.0005).
Since transcription levels do not always accurately
reflect the amount of translated protein, western blots were
AB
*
col6a1
CD
Percentage fibrosis
WT KO
ns
WT
KO
*
0
1
2
3
4
0
50
100
150
WT KO
40
30
20
6
4
2
0
WT
KO
2
-Δct
versus Hprt1 (X100)
2
-Δct
versus Hprt1 (X100)
-subunits -subunits
Fig. 2 Kcna1-null mice exhibit
minimal Kv1.x channel
remodeling and atrial fibrosis.
aReal-time PCR expression
analysis of Kv1.x potassium
channel a- and b-subunit genes
in atria from KO and control
WT mice (n=5 mice per
genotype). bReal-time PCR
analysis of col6a1 mRNA levels
(n=5 mice per genotype).
cRepresentative samples of
atria from WT and KO animals
stained with Masson’s
trichrome to visualize fibrosis
which appears bluish. Images
were chosen out of sections
from 5 WT mice and 5 KO
mice. dQuantification of the
percentage of atrial fibrosis
between genotypes. *P\0.05;
ns not significant
47 Page 8 of 15 Basic Res Cardiol (2015) 110:47
123
performed to measure Kv1.1 subunit abundance in patients
and controls. Immunoblots of atrial tissue revealed the
presence of detectable amounts of Kv1.1 protein in human
heart for the first time (Fig. 4c, d). Quantification of Kv1.1
subunit levels revealed no change in Kv1.1 protein levels
in pAF patients (P =0.804) but an unexpected and sig-
nificant 75 % increase in Kv1.1 protein levels in cAF
patients compared to SR controls (Fig. 4c, d; P\0.05).
Kv1.1 protein appeared similarly increased in cAF patients
with and without valvular heart disease (Supplementary
Figure 1). To verify that human Kv1.1 protein expression
was associated with cardiomyocytes and not with other cell
types, immunocytochemistry of isolated human atrial
myocytes was performed revealing significant myocyte-
Fig. 3 Kcna1 expression in
mouse cardiomyocytes. aRT-
PCR detection of Kcna1 mRNA
in isolated atrial (A) and
ventricular (V) myocytes from
three mice labeled 1-3. Brain
tissue (B) was used as a positive
control, while no cDNA (N) was
used as a negative control.
bQuantitative RT-PCR shows
that Kcna1 mRNA levels
(normalized to Rpl7) are about
tenfold more abundant in atrial
myocytes than in ventricular
(n =3 mice; P=0.07, paired
ttest). Immunostaining of
isolated atrial (c) and ventricular
(d) myocytes isolated from
adult mice and co-stained with
antibodies against Kv1.1 and
Junctophilin-2 (Jph2), a t-tubule
protein. The gain was increased
in the ventricular images
relative to the atrial images to
allow visualization of Kv1.1
immunoreactivity, which is
much less intense in ventricular
myocytes. The boxed regions in
the overlay images are shown
magnified 500 % in the bottom
panels. Representative images
were chosen out of 15 cells (7
atrial and 8 ventricular
myocytes) from 2 WT mice and
8 cells (5 atrial and 3 ventricular
myocytes) from 1 KO mouse.
Scale bars 20 lm
Basic Res Cardiol (2015) 110:47 Page 9 of 15 47
123
specific Kv1.1 immunoreactivity (Fig. 5). The substantial
upregulation of Kv1.1 subunits in the atria of cAF patients,
but not pAF, suggests that the channels undergo expression
remodeling that may contribute to AF pathophysiology and
be a potential determinant of the transition to maintained
AF.
Kv1.1 subunits form functional channels in human
atrial myocytes
Having detected Kv1.1 subunits in human atria, patch-
clamp recordings were performed utilizing a Kv1.1-speci-
fic inhibitor, dendrotoxin-K (DTX-K), to determine whe-
ther the channels make a functional contribution to human
cardiac currents [31]. Following enzymatic isolation of
human myocytes from right atrial appendages of patients in
SR, whole-cell voltage clamp recordings were performed
to measure outward membrane currents using a voltage
step protocol composed of brief one-second depolarizations
from -60 mV to ?50 mV at 0.1 Hz to activate outward
K
?
currents (Fig. 6a). In the presence of DTX-K to
selectively block Kv1.1 subunits, the peak and late outward
current amplitudes were significantly reduced by about 20
and 10 %, respectively (Fig. 6b, c; P\0.01 and 0.05,
respectively). This DTX-K-sensitive component of the
outward current is indicative of a small but significant
contribution by Kv1.1-containing channels to the total
repolarizing K
?
current in human atrial myocytes. Thus,
Kv1.1 channels are not only present in human heart, but
they are also functional and contribute to atrial
repolarization.
Kv1.1-associated currents are increased in cAF
patients
To determine if the observed cAF-associated increase in
Kv1.1 protein levels corresponds with an increase in
Kv1.1-associated currents, the DTX-K-sensitive compo-
nent was then measured in atrial myocytes isolated from
patients with cAF. Membrane capacitance was not signif-
icantly different between SR and cAF myocytes suggesting
similar cell sizes in each group (P=0.59; Supplementary
Figure 2). Myocytes from patients with cAF showed a
slight reduction in K
?
-current amplitudes, which did not
reach the level of statistical significance (Fig. 6a). How-
ever, both the peak and late components of the DTX-K-
sensitive current showed a significant two- to three-fold
increase compared to SR (Fig. 6b–d; P\0.05 and 0.01 at
peak and late current levels, respectively). To further
characterize our patient cohort, I
Ca,L
was measured in
myocytes from SR and cAF patients as a major hallmark of
electrical remodeling. Consistent with previous reports,
I
Ca,L
was significantly reduced by about 50 % in cAF
myocytes compared to SR (Supplementary Figure 3) [46].
The substantial increase in DTX-K-sensitive current in cAF
patients, coupled with increased KCNA1 expression,
0
5
10
15
76
9
SR pAF
0.0
0.5
1.0
1.5
2.0
2.5
15 14
Relative to SR
(KCNA1/HPRT1)
500 bp
1500 bp
KCNA1
A
ns
CD
*
Kv1.1 - 56 kDa
GAPDH - 34 kDa
SR SR cAF cAF
Relative to SR
(norm. to GAPDH)
SR cAF
0.0
0.5
1.0
1.5
2.0
2.5
911
*
Kv1.1 - 56 kDa
GAPDH - 34 kDa
SR SR pAF pAF
R
S
o
t
e
vi
tal
e
R
)
H
D
P
AGo
t
.m
ron(
ns
SR pAF cAF
B
Fig. 4 KCNA1 mRNA and
protein expression in human
atria. aRT-PCR detection of
KCNA1 mRNA in human atria.
bmRNA levels (mean ±SEM)
of KCNA1 in patients with
chronic AF (cAF) and
paroxysmal AF (pAF) relative
to sinus rhythm (SR).
Representative Western blots of
Kv1.1 protein with
corresponding densitometric
quantification (mean ±SEM)
of protein levels in patients with
pAF (c) and cAF (d) relative to
SR. *P\0.05 vs. SR; ns not
significant. Numbers in bars
indicate atrial samples
47 Page 10 of 15 Basic Res Cardiol (2015) 110:47
123
suggests that Kv1.1 channels undergo molecular and
electrical remodeling, which may contribute to the path-
omechanisms of persistent AF.
Discussion
Traditionally, Kv1.1 channels have been regarded as brain-
specific channels with little to no cardiac expression but
our evidence suggests that this paradigm needs to be
reconsidered. Here RT-PCR, western blotting, and
immunocytochemistry demonstrate the presence of Kv1.1
channels in human atria; the first time they have been
reported in human heart. Prior studies found Kv1.1 tran-
scripts in mouse and rat hearts using Northern blotting but
the abundance was so low that they were thought to result
from sample contamination by neural tissues [18,30].
More recently, newer RT-PCR-based methods with higher
sensitivity reproducibly detected Kv1.1 transcripts in
mouse heart, especially in nodal regions where the gene
appears to be expressed at relatively high levels [11,12,14,
20]. This work verifies the presence of Kv1.1 mRNA in
mouse atria, albeit at apparently low abundance. Impor-
tantly, Kv1.1 mRNA was detected in isolated mouse car-
diomyocytes for the first time in this study, demonstrating
that Kv1.1 expression in cardiac tissue cannot be solely
attributed to contamination by neural cells. Interestingly,
Kv1.1 transcripts showed higher expression in atrial myo-
cytes than ventricular, similar to what has been reported for
Kv1.5 subunits [39]. Furthermore, at the protein level, this
study provides the first evidence of Kv1.1 immunoreac-
tivity in individual cardiomyocytes. In mice, the Kv1.1-
positive labeling in atrial myocytes appeared predomi-
nantly intracellular, whereas the weaker staining in ven-
tricular myocytes showed clustering in a pattern consistent
with transverse tubules, as revealed by co-labeling with the
t-tubule marker protein Junctophilin-2. The Kv1.1
immunoreactivity in human atrial myocytes was similar to
that seen in mice with a predominantly intracellular
staining pattern, as well as striations consistent with
localization at Z-lines. The mostly intracellular Kv1.1
staining pattern in atrial myocytes may reflect a relative
abundance of the channel in intracellular pools awaiting
membrane insertion in response to some physiological
trigger. For example, atrial Kv1.5 channels are dynamically
recruited to the membrane from submembranous pools in
response to high sheer stress or cholesterol depletion [1,3].
However, further experimentation will be required to
determine whether Kv1.1 channels undergo similar traf-
ficking mechanisms.
Our identification of DTX-K-sensitive outward currents
in human atrial myocytes suggests that Kv1.1 channels
make a previously unrecognized contribution to membrane
repolarization during the atrial action potential. Biochem-
ical and electrophysiological studies have repeatedly
demonstrated the exquisitely strict specificity of DTX-K
for inhibition of only Kv1.1 subunits [31,48,49]. The two
primary voltage-gated potassium currents in human atrial
myocytes are the fast transient outward current (I
to,f
) and
the ultra-rapid delayed rectifier potassium current (I
Kur
),
which are encoded by Kv4.3 and Kv1.5 a-subunits,
respectively. Since DTX-K reduced the peak and late
outward currents, we predict that Kv1.1 channels make a
small but significant contribution to I
to,f
and I
Kur
. Of note,
Fig. 5 Kv1.1 immunoreactivity in isolated human atrial cardiomy-
ocytes. Representative confocal images of Kv1.1-positive immuno-
staining (a,b) and a negative control (c). Kv1.1 immunoreactivity
showed a strong fluorescent signal throughout the cell and a tight
striated pattern which overlapped with the Z-line. In negative
controls, primary antibody was omitted and cells were labeled only
with the secondary antibody revealing an absence of nonspecific,
background staining
Basic Res Cardiol (2015) 110:47 Page 11 of 15 47
123
human atrial myocytes exhibit a substantial late outward
I
Kur
current component of unknown molecular origin and
composition that is characterized as being non-inactivating,
insensitive to the Kv1.5-blocker AVE0118, and increased
in cAF myocytes relative to SR [6,41]. The DTX-K-sen-
sitive late currents recorded here fit the profile of this
unidentified non-inactivating, AVE0118-insensitive current
component of I
Kur
, suggesting that it may be encoded by
Kv1.1 channels. In addition to I
to,f
and I
Kur
, human atrial
myocytes also exhibit a rapidly activating and inactivating
current (I
Kr
) and a slowly activating current (I
Ks
), mediated
by hERG1 and KCNQ1 channels, respectively, but they are
0
1
2
3
4
DTX-K sensitive
current (pA/pF)
"late""peak"
*
**
0
2
4
6
8
10
***
***
"late""peak"
0
2
4
6
8
10
**
Current amplitude
(pA/pF)
"late""peak"
*
500 ms
0 pA/pF
5 pA/pF
500 ms
0 pA/pF
5 pA/pF
DTX-K sensitive current
A
B
C
500 ms
0 pA/pF
4 pA/pF
DTX-K
(10 nM)
Control
500 ms
0 pA/pF
4 pA/pF
SR cAF
DTX-K
(10 nM)
Control
”peak“
“late“ ”peak“
“late“
-60 mV
50 mV
500 ms
f = 0.1 Hz
n=9/3 n=11/3
Control
DTX-K
SR (n=9/3)
cAF (n=11/3)
SR cAF
D
Fig. 6 Dendrotoxin-K sensitive
outward current in isolated
human atrial myocytes.
aAveraged current traces
recorded from human atrial
myocytes from patients in sinus
rhythm (SR) or chronic AF
(cAF). Current traces show
control measurements at
baseline followed by application
of 10 nmol/L dendrotoxin-K
(DTX-K) to specifically block
Kv1.1 channels. The voltage
step protocol used for
recordings is shown at the top.
bTraces show the averaged
current difference representing
the dendrotoxin-K sensitive
component. cAmplitudes of the
peak and late phases of the
outward K
?
currents
(mean ±SEM) before (control)
and after application of DTX-K
in atrial myocytes from SR and
cAF patients. dAmplitudes of
the peak and late phases of the
DTX-K sensitive current
difference (mean ±SEM) in
atrial myocytes from SR and
cAF patients. *P\0.05,
**P\0.01, ***P\0.001 vs.
corresponding means under
control conditions using
Student’s paired (c) and
unpaired (d) t test, respectively.
The nvalues indicate
myocyte/patient numbers
47 Page 12 of 15 Basic Res Cardiol (2015) 110:47
123
much smaller outward potassium currents relative to I
to,f
and I
Kur
[2,24,33,34,40]. Whether Kv1.1 channels
contribute to I
Kr
or I
Ks
cannot be inferred from this work,
since the myocyte isolation protocol largely destroys the
I
Kr
current and the voltage step duration is too short to
accurately discriminate classical I
Ks
. In summary, human
atrial myocytes exhibit significant DTX-K-sensitive peak
and late outward currents that may represent a previously
unrecognized contribution by Kv1.1 channels to I
to,f
and
I
Kur
.
Elucidation of the subunit stoichiometry of the func-
tional cardiac Kv1.1 channel may help resolve the identity
and role of the Kv1.1-associated cardiac current in the
atrial action potential. The kinetics and gating properties of
Kv1-mediated currents are determined by their associated
a- and b-subunit composition. For example, Kv1.4 a-sub-
units and Kvb1 accessory subunits confer fast N-type
inactivation via the presence of ball domains, whereas
Kv1.6 a-subunits possess N-type inactivation prevention
(NIP) domains that can neutralize inactivation gating [27,
32,38]. Studies in brain show that Kv1.1 a-subunits
preferentially form functional heterotetrameric assemblies
with Kv1.2 or Kv1.4 and sometimes Kv1.6 [49]. Future
biochemical analyses will be required to determine which
Kv1 a- and b-subunit combinations exist in heart, and what
their respective functional roles might be.
The significant increase in DTX-K-sensitive currents in
cAF myocytes with concomitant upregulation of Kv1.1
protein levels suggests that Kv1.1 channels may contribute to
electrical remodeling in cAF. The transition from paroxys-
mal to persistent AF is facilitated by atrial remodeling of ion
channels at the level of expression and electrophysiological
properties [9,13,23,42,45]. These AF-related electrical
adaptations generally promote shortening of the atrial action
potential and blunting of the atrial effective refractory per-
iod, which increases vulnerability to AF by providing a pro-
arrhythmogenic substrate prone to premature beats [9,23]. In
AF patients, Kv4.3 channel mRNA and protein are down-
regulated leading to a 60 % decrease in I
to
, which is thought
to indirectly increase the upstroke velocity of the atrial action
potential leading to augmented wave propagation that pro-
motes maintenance of AF [5,51]. Remodeling has also been
reported for the atrial voltage-gated K
?
channels hERG1,
Kv1.5 and KCNQ1, but their expression changes have either
been inconsistent or not correlated with corresponding
changes in atrial currents [23]. The concurrent increase in
Kv1.1 protein expression and Kv1.1-mediated outward
currents provides one of the first examples of cAF-associated
upregulation remodeling of a voltage-gated potassium
channel. Interestingly, we did not find corresponding
increases in KCNA1 mRNA levels in cAF atria suggesting
the influence of post-transcriptional modifications similar to
Kv1.5 channels which also exhibit a discordance between
mRNA and protein levels in cAF patients [4,5]. The aug-
mentation of Kv1.1 protein expression and currents is con-
sistent with the reduced action potential durations
characteristic of AF, but whether these are primary causative
changes or secondary compensations remains to be tested.
The increased incidence of inducible AF in Kv1.1-de-
ficient mice suggests that Kcna1 may be a new candidate
gene for AF susceptibility; however, the overexpression of
Kv1.1 channels in cAF patients appears contradictory to
this notion. Some of this discrepancy could be related to
inter-species differences in atrial physiology and channel
expression between mice and humans [17]. Loss-of-func-
tion mutations in the related voltage-gated potassium
channel KCNA5 unexpectedly lead to AF by an atypical
mechanism involving decreased I
Kur
which leads to a
predisposition for atrial action potential prolongation and
early after-depolarizations [25,52]. Kv1.1-deficiency may
cause similar complex AF-promoting alterations in atrial
function; however, in humans KCNA1 gene variants have
yet to be associated with AF. Determination of whether the
upregulation of Kv1.1 channels in cAF patients is causative
for AF or a compensatory mechanism is necessary to
resolve the role of Kv1.1 channels in AF. If it is com-
pensatory, Kv1.1 currents would be expected to oppose AF
and their absence would be expected to promote AF as seen
in our mouse model. Additional studies will be required to
dissect the exact contribution of Kv1.1 currents to action
potential duration and AF susceptibility in mice and
humans, especially given the complex relationship between
modification of outward potassium currents and action
potential morphology [7].
Acknowledgments The authors thank Ramona Nagel and Katrin
Kupser for excellent technical assistance. This work was supported by
grants from the National Institutes of Health (HL107641 to EG;
HL089598, HL091947, and HL117641 to X.H.T.W.; NS076916 to
J.L.N.), the Muscular Dystrophy Association (X.H.T.W.), American
Heart Association (13EIA14560061 to X.H.T.W.), the Deutsche
Forschungsgemeinschaft (Do 769/1-1-3 to D.D.), the German Federal
Ministry of Education and Research through DZHK (German Centre
for Cardiovascular Research to D.D.), the European Union through
the European Network for Translational Research in Atrial Fibrilla-
tion (EUTRAF, FP7-HEALTH-2010, large-scale integrating project,
Proposal No. 261057 to D.D), and Fondation Leducq (‘Alliance for
CaMKII Signaling in Heart’ to X.H.T.W. and ‘European North-
American Atrial Fibrillation Research Alliance’ to D.D.).
Compliance with ethical standards
Conflicts of interest None.
References
1. Balse E, El-Haou S, Dillanian G, Dauphin A, Eldstrom J, Fedida
D, Coulombe A, Hatem SN (2009) Cholesterol modulates the
recruitment of Kv1.5 channels from Rab11-associated recycling
Basic Res Cardiol (2015) 110:47 Page 13 of 15 47
123
endosome in native atrial myocytes. Proc Natl Acad Sci
106:14681–14686. doi:10.1073/pnas.0902809106
2. Barhanin J, Lesage F, Guillemare E, Fink M, Lazdunski M,
Romey G (1996) K(V)LQT1 and lsK (minK) proteins associate to
form the I(Ks) cardiac potassium current. Nature 384:78–80.
doi:10.1038/384078a0
3. Boycott HE, Barbier CSM, Eichel CA, Costa KD, Martins RP,
Louault F, Dilanian G, Coulombe A, Hatem SN, Balse E (2013)
Shear stress triggers insertion of voltage-gated potassium chan-
nels from intracellular compartments in atrial myocytes. Proc
Natl Acad Sci 110:E3955–E3964. doi:10.1073/pnas.1309896110
4. Brundel BJ, Van Gelder IC, Henning RH, Tieleman RG,
Tuinenburg AE, Wietses M, Grandjean JG, Van Gilst WH, Crijns
HJ (2001) Ion channel remodeling is related to intraoperative
atrial effective refractory periods in patients with paroxysmal and
persistent atrial fibrillation. Circulation 103:684–690. doi:10.
1161/01.CIR.103.5.684
5. Brundel BJ, Van Gelder IC, Henning RH, Tuinenburg AE,
Wietses M, Grandjean JG, Wilde AA, Van Gilst WH, Crijns HJ
(2001) Alterations in potassium channel gene expression in atria
of patients with persistent and paroxysmal atrial fibrillation:
differential regulation of protein and mRNA levels for K?
channels. J Am Coll Cardiol 37:926–932. doi:10.1016/S0735-
1097(00)01195-5
6. Christ T, Wettwer E, Voigt N, Ha
´la O, Radicke S, Matschke K,
Va
´rro A, Dobrev D, Ravens U (2008) Pathology-specific effects
of the IKur/Ito/IK, ACh blocker AVE0118 on ion channels in
human chronic atrial fibrillation. Br J Pharmacol 154:1619–1630.
doi:10.1038/bjp.2008.209
7. Courtemanche M, Ramirez RJ, Nattel S (1999) Ionic targets for
drug therapy and atrial fibrillation-induced electrical remodeling:
insights from a mathematical model. Cardiovasc Res 42:477–489.
doi:10.1016/S0008-6363(99)00034-6
8. Dahab GM, Kheriza MM, El-Beltagi HM, Fouda A-MM, El-Din
OAS (2004) Digital quantificationof fibrosis in liver biopsy sections:
description of a new method by Photoshop software. J Gastroenterol
Hepatol 19:78–85. doi:10.1111/j.1440-1746.2004.03183.x
9. Dobrev D, Ravens U (2003) Remodeling of cardiomyocyte ion
channels in human atrial fibrillation. Basic Res Cardiol
98:137–148. doi:10.1007/s00395-003-0409-8
10. Glasscock E, Qian J, Kole MJ, Noebels JL (2012) Transcom-
partmental reversal of single fibre hyperexcitability in juxta-
paranodal Kv1.1-deficient vagus nerve axons by activation of
nodal KCNQ channels. J Physiol 590:3913–3926. doi:10.1113/
jphysiol.2012.235606
11. Glasscock E, Yoo JW, Chen TT, Klassen TL, Noebels JL (2010)
Kv1.1 potassium channel deficiency reveals brain-driven cardiac
dysfunction as a candidate mechanism for sudden unexplained
death in epilepsy. J Neurosci 30:5167–5175. doi:10.1523/
JNEUROSCI.5591-09.2010
12. Harrell MD, Harbi S, Hoffman JF, Zavadil J, Coetzee WA (2007)
Large-scale analysis of ion channel gene expression in the mouse
heart during perinatal development. Physiol Genomics
28:273–283. doi:10.1152/physiolgenomics.00163.2006
13. Heijman J, Dobrev D (2015) Irregular rhythm and atrial meta-
bolism are key for the evolution of proarrhythmic atrial remod-
eling in atrial fibrillation. Basic Res Cardiol 110:498. doi:10.
1007/s00395-015-0498-1
14. Leoni A-L, Marionneau C, Demolombe S, Le Bouter S, Mangoni
ME, Escande D, Charpentier F (2005) Chronic heart rate reduc-
tion remodels ion channel transcripts in the mouse sinoatrial node
but not in the ventricle. Physiol Genomics 24:4–12. doi:10.1152/
physiolgenomics.00161.2005
15. Li N, Wang T, Wang W, Cutler MJ, Wang Q, Voigt N, Rosen-
baum DS, Dobrev D, Wehrens XHT (2012) Inhibition of CaMKII
phosphorylation of RyR2 prevents induction of atrial fibrillation
in FKBP12.6 knockout mice. Circ Res 110:465–470. doi:10.
1161/CIRCRESAHA.111.253229
16. Livak KJ, Schmittgen TD (2001) Analysis of relative gene
expression data using real-time quantitative PCR and the
2(-Delta Delta C(T)) Method. Methods San Diego Calif
25:402–408. doi:10.1006/meth.2001.1262
17. London B (2001) Cardiac arrhythmias: from (transgenic) mice to
men. J Cardiovasc Electrophysiol 12:1089–1091. doi:10.1046/j.
1540-8167.2001.01089.x
18. London B, Guo W, Pan Xh, Lee JS, Shusterman V, Rocco CJ,
Logothetis DA, Nerbonne JM, Hill JA (2001) Targeted replace-
ment of KV1.5 in the mouse leads to loss of the 4-aminopyridine-
sensitive component of I(K, slow) and resistance to drug-induced
qt prolongation. Circ Res 88:940–946. doi:10.1161/hh0901.
090929
19. Mahida S, Lubitz SA, Rienstra M, Milan DJ, Ellinor PT (2011)
Monogenic atrial fibrillation as pathophysiological paradigms.
Cardiovasc Res 89:692–700. doi:10.1093/cvr/cvq381
20. Marionneau C, Couette B, Liu J, Li H, Mangoni ME, Nargeot J,
Lei M, Escande D, Demolombe S (2005) Specific pattern of ionic
channel gene expression associated with pacemaker activity in
the mouse heart. J Physiol 562:223–234. doi:10.1113/jphysiol.
2004.074047
21. Nattel S (2002) New ideas about atrial fibrillation 50 years on.
Nature 415:219–226. doi:10.1038/415219a
22. Nattel S, Burstein B, Dobrev D (2008) Atrial remodeling and
atrial fibrillation: mechanisms and implications. Circ Arrhythm
Electrophysiol 1:62–73. doi:10.1161/CIRCEP.107.754564
23. Nattel S, Maguy A, Le Bouter S, Yeh Y-H (2007) Arrhythmo-
genic ion-channel remodeling in the heart: heart failure,
myocardial infarction, and atrial fibrillation. Physiol Rev
87:425–456. doi:10.1152/physrev.00014.2006
24. Nerbonne JM, Kass RS (2005) Molecular physiology of cardiac
repolarization. Physiol Rev 85:1205–1253. doi:10.1152/physrev.
00002.2005
25. Olson TM, Alekseev AE, Liu XK, Park S, Zingman LV,
Bienengraeber M, Sattiraju S, Ballew JD, Jahangir A, Terzic A
(2006) Kv1.5 channelopathy due to KCNA5 loss-of-function
mutation causes human atrial fibrillation. Hum Mol Genet
15:2185–2191. doi:10.1093/hmg/ddl143
26. Van Oort RJ, Garbino A, Wang W, Dixit SS, Landstrom AP,
Gaur N, De Almeida AC, Skapura DG, Rudy Y, Burns AR,
Ackerman MJ, Wehrens XHT (2011) Disrupted junctional
membrane complexes and hyperactive ryanodine receptors after
acute junctophilin knockdown in mice. Circulation 123:979–988.
doi:10.1161/CIRCULATIONAHA.110.006437
27. Rettig J, Heinemann SH, Wunder F, Lorra C, Parcej DN, Dolly
JO, Pongs O (1994) Inactivation properties of voltage-gated K?
channels altered by presence of beta-subunit. Nature
369:289–294. doi:10.1038/369289a0
28. Reynolds JO, Chiang DY, Wang W, Beavers DL, Dixit SS,
Skapura DG, Landstrom AP, Song L-S, Ackerman MJ, Wehrens
XHT (2013) Junctophilin-2 is necessary for T-tubule maturation
during mouse heart development. Cardiovasc Res 100:44–53.
doi:10.1093/cvr/cvt133
29. Robbins CA, Tempel BL (2012) Kv1.1 and Kv1.2: similar
channels, different seizure models. Epilepsia 53(Suppl
1):134–141. doi:10.1111/j.1528-1167.2012.03484.x
30. Roberds SL, Tamkun MM (1991) Cloning and tissue-specific
expression of five voltage-gated potassium channel cDNAs
expressed in rat heart. Proc Natl Acad Sci USA 88:1798–1802.
doi:10.1073/pnas.88.5.1798
31. Robertson B, Owen D, Stow J, Butler C, Newland C (1996)
Novel effects of dendrotoxin homologues on subtypes of mam-
malian Kv1 potassium channels expressed in Xenopus oocytes.
FEBS Lett 383:26–30. doi:10.1016/0014-5793(96)00211-6
47 Page 14 of 15 Basic Res Cardiol (2015) 110:47
123
32. Roeper J, Sewing S, Zhang Y, Sommer T, Wanner SG, Pongs O
(1998) NIP domain prevents N-type inactivation in voltage-gated
potassium channels. Nature 391:390–393. doi:10.1038/34916
33. Sanguinetti MC, Curran ME, Zou A, Shen J, Spector PS,
Atkinson DL, Keating MT (1996) Coassembly of K(V)LQT1 and
minK (IsK) proteins to form cardiac I(Ks) potassium channel.
Nature 384:80–83. doi:10.1038/384080a0
34. Sanguinetti MC, Jiang C, Curran ME, Keating MT (1995) A
mechanistic link between an inherited and an acquired cardiac
arrhythmia: HERG encodes the IKr potassium channel. Cell
81:299–307
35. Sinner MF, Ellinor PT, Meitinger T, Benjamin EJ, Ka
¨a
¨b S (2011)
Genome-wide association studies of atrial fibrillation: past, pre-
sent, and future. Cardiovasc Res 89:701–709. doi:10.1093/cvr/
cvr001
36. Smart SL, Lopantsev V, Zhang CL, Robbins CA, Wang H, Chiu
SY, Schwartzkroin PA, Messing A, Tempel BL (1998) Deletion
of the K(V)1.1 potassium channel causes epilepsy in mice.
Neuron 20:809–819
37. Sood S, Chelu MG, van Oort RJ, Skapura D, Santonastasi M,
Dobrev D, Wehrens XHT (2008) Intracellular calcium leak due to
FKBP12.6 deficiency in mice facilitates the inducibility of atrial
fibrillation. Heart Rhythm 5:1047–1054. doi:10.1016/j.hrthm.
2008.03.030
38. Stu
¨hmer W, Ruppersberg JP, Schro
¨ter KH, Sakmann B, Stocker
M, Giese KP, Perschke A, Baumann A, Pongs O (1989) Molec-
ular basis of functional diversity of voltage-gated potassium
channels in mammalian brain. EMBO J 8:3235–3244
39. Tamkun MM, Knoth KM, Walbridge JA, Kroemer H, Roden
DM, Glover DM (1991) Molecular cloning and characterization
of two voltage-gated K?channel cDNAs from human ventricle.
FASEB J Off Publ Fed Am Soc Exp Biol 5:331–337
40. Trudeau MC, Warmke JW, Ganetzky B, Robertson GA (1995)
HERG, a human inward rectifier in the voltage-gated potassium
channel family. Science 269:92–95
41. Turnow K, Metzner K, Cotella D, Morales MJ, Schaefer M,
Christ T, Ravens U, Wettwer E, Ka
¨mmerer S (2015) Interaction
of DPP10a with Kv4.3 channel complex results in a sustained
current component of human transient outward current Ito. Basic
Res Cardiol 110:5. doi:10.1007/s00395-014-0457-2
42. Voigt N, Heijman J, Wang Q, Chiang DY, Li N, Karck M,
Wehrens XHT, Nattel S, Dobrev D (2014) Cellular and molecular
mechanisms of atrial arrhythmogenesis in patients with parox-
ysmal atrial fibrillation. Circulation 129:145–156. doi:10.1161/
CIRCULATIONAHA.113.006641
43. Voigt N, Li N, Wang Q, Wang W, Trafford AW, Abu-Taha I, Sun
Q, Wieland T, Ravens U, Nattel S, Wehrens XHT, Dobrev D
(2012) Enhanced sarcoplasmic reticulum Ca2?leak and
increased Na?-Ca2?exchanger function underlie delayed after
depolarizations in patients with chronic atrial fibrillation. Circu-
lation 125:2059–2070. doi:10.1161/CIRCULATIONAHA.111.
067306
44. Voigt N, Makary S, NattelS, Dobrev D (2010) Voltage-clamp-based
methods for the detection of constitutively active acetylcholine-
gated I(K, ACh) channels in the diseased heart. Methods Enzymol
484:653–675. doi:10.1016/B978-0-12-381298-8.00032-0
45. Voigt N, Trausch A, Knaut M, Matschke K, Varro
´A, Van
Wagoner DR, Nattel S, Ravens U, Dobrev D (2010) Left-to-right
atrial inward rectifier potassium current gradients in patients with
paroxysmal versus chronic atrial fibrillation. Circ Arrhythm
Electrophysiol 3:472–480. doi:10.1161/CIRCEP.110.954636
46. Van Wagoner DR, Pond AL, Lamorgese M, Rossie SS, McCarthy
PM, Nerbonne JM (1999) Atrial L-type Ca2?currents and
human atrial fibrillation. Circ Res 85:428–436
47. Wakili R, Voigt N, Ka
¨a
¨b S, Dobrev D, Nattel S (2011) Recent
advances in the molecular pathophysiology of atrial fibrillation.
J Clin Invest 121:2955–2968. doi:10.1172/JCI46315
48. Wang FC, Bell N, Reid P, Smith LA, McIntosh P, Robertson B,
Dolly JO (1999) Identification of residues in dendrotoxin K
responsible for its discrimination between neuronal K?channels
containing Kv1.1 and 1.2 alpha subunits. Eur J Biochem FEBS
263:222–229
49. Wang FC, Parcej DN, Dolly JO (1999) Alpha subunit composi-
tions of Kv1.1-containing K?channel subtypes fractionated from
rat brain using dendrotoxins. Eur J Biochem FEBS 263:230–237
50. Wijffels MC, Kirchhof CJ, Dorland R, Allessie MA (1995) Atrial
fibrillation begets atrial fibrillation. A study in awake chronically
instrumented goats. Circulation 92:1954–1968
51. Workman AJ, Kane KA, Rankin AC (2001) The contribution of
ionic currents to changes in refractoriness of human atrial myo-
cytes associated with chronic atrial fibrillation. Cardiovasc Res
52:226–235
52. Yang Y, Li J, Lin X, Yang Y, Hong K, Wang L, Liu J, Li L, Yan
D, Liang D, Xiao J, Jin H, Wu J, Zhang Y, Chen Y-H (2009)
Novel KCNA5 loss-of-function mutations responsible for atrial
fibrillation. J Hum Genet 54:277–283. doi:10.1038/jhg.2009.26
Basic Res Cardiol (2015) 110:47 Page 15 of 15 47
123

Supplementary resource (1)

... Devisnki et al. (2016), 1 Chalhal et al. (2020),6 Coll et al. (2015),35 Thom et al. (2018),36 Bagnall et al. (2017),37 Goldman et al. (2016),38 Glasscock et al. (2015),39 Trosclair et al. (2020), 40 Trosclair et al. (2021), 41 Glasscock et al. (2019), 42 Frasier et al. (2016). 43 AV, atrioventricular; CPVT, Catecholaminergic polymorphic ventricular tachycardia; VF, Ventricular fibrillation. ...
... Devisnki et al. (2016), 1 Chalhal et al. (2020),6 Coll et al. (2015),35 Thom et al. (2018),36 Bagnall et al. (2017),37 Goldman et al. (2016),38 Glasscock et al. (2015),39 Trosclair et al. (2020), 40 Trosclair et al. (2021), 41 Glasscock et al. (2019), 42 Frasier et al. (2016). 43 AV, atrioventricular; CPVT, Catecholaminergic polymorphic ventricular tachycardia; VF, Ventricular fibrillation. ...
Article
Full-text available
The influence of the central nervous system and autonomic system on cardiac activity is being intensively studied, as it contributes to the high rate of cardiologic comorbidities observed in people with epilepsy. Indeed, neuroanatomic connections between the brain and the heart provide links that allow cardiac arrhythmias to occur in response to brain activation, have been shown to produce arrhythmia both experimentally and clinically. Moreover, seizures may induce a variety of transient cardiac effects, which include changes in heart rate, heart rate variability, arrhythmias, asystole, and other ECG abnormalities, and can trigger the development of Takotsubo syndrome. People with epilepsy are at a higher risk of death than the general population, and sudden unexpected death in epilepsy (SUDEP) is the most important direct epilepsy‐related cause of death. Although the cause of SUDEP is still unknown, cardiac abnormalities during and between seizures could play a significant role in its pathogenesis, as highlighted by studies on animal models of SUDEP and registration of SUDEP events. Recently, genetic mutations in genes co‐expressed in the heart and brain, which may result in epilepsy and cardiac comorbidity/increased risk for SUDEP, have been described. Recognition and a better understanding of brain–heart interactions, together with new advances in sequencing techniques, may provide new insights into future novel therapies and help in the prevention of cardiac dysfunction and sudden death in epileptic individuals.
... The pathogenesis of AF is multifaceted, involving abnormalities in the conduction system, the effects of the autonomic nervous system, and abnormal ionic current metabolism in cardiomyocytes (3)(4)(5)(6). Current research focuses on changes in ion channels, extracellular matrix, and intracellular signaling pathways. ...
Article
Full-text available
Background Atrial fibrillation (AF) is a common cardiac arrhythmia that is associated with obesity and obstructive sleep apnea syndrome (OSA). Obesity and OSA may increase the risk of AF by affecting cardiovascular health. Methods The study used the Mendelian randomization (MR) approach, combined with two-sample and multivariable analyses, to assess the relationships between obesity, OSA, and AF. The study utilized GWAS data and applied various statistical methods for the analysis. Results The study found that obesity increased the risk of OSA, which in turn significantly increased the risk of AF. Through mediating MR analysis, it was found that OSA played a certain role in the causal relationship between obesity and AF, with about 6.4% of the risk of AF being mediated by OSA. Conclusion This study highlights the relationships among obesity, OSA, and AF, providing useful guidance for future clinical researches.
... When a dominantnegative fragment of KCNA1 was expressed, mice developed LQT and spontaneous ventricular arrhythmias 283 . Interestingly, it was later shown by Glasscock et al. 284 that KCNA1 is preferentially expressed (≈tenfold higher) in atria over ventricles. Programmed electrical stimulation (PES) induced AF in KCNA1 knockout mice but did not lead to any ventricular arrhythmias and no differences in QT interval were observed. ...
Article
Full-text available
Cardiac rhythm regulated by micro-macroscopic structures of heart. Pacemaker abnormalities or disruptions in electrical conduction, lead to arrhythmic disorders may be benign, typical, threatening, ultimately fatal, occurs in clinical practice, patients on digitalis, anaesthesia or acute myocardial infarction. Both traditional and genetic animal models are: In-vitro: Isolated ventricular Myocytes, Guinea pig papillary muscles, Patch-Clamp Experiments, Porcine Atrial Myocytes, Guinea pig ventricular myocytes, Guinea pig papillary muscle: action potential and refractory period, Langendorff technique, Arrhythmia by acetylcholine or potassium. Acquired arrhythmia disorders: Transverse Aortic Constriction, Myocardial Ischemia, Complete Heart Block and AV Node Ablation, Chronic Tachypacing, Inflammation, Metabolic and Drug-Induced Arrhythmia. In-Vivo: Chemically induced arrhythmia: Aconitine antagonism, Digoxin-induced arrhythmia, Strophanthin/ouabain-induced arrhythmia, Adrenaline-induced arrhythmia, and Calcium-induced arrhythmia. Electrically induced arrhythmia: Ventricular fibrillation electrical threshold, Arrhythmia through programmed electrical stimulation, sudden coronary death in dogs, Exercise ventricular fibrillation. Genetic Arrhythmia: Channelopathies, Calcium Release Deficiency Syndrome, Long QT Syndrome, Short QT Syndrome, Brugada Syndrome. Genetic with Structural Heart Disease: Arrhythmogenic Right Ventricular Cardiomyopathy/Dysplasia, Dilated Cardiomyopathy, Hypertrophic Cardiomyopathy, Atrial Fibrillation, Sick Sinus Syndrome, Atrioventricular Block, Preexcitation Syndrome. Arrhythmia in Pluripotent Stem Cell Cardiomyocytes. Conclusion: Both traditional and genetic, experimental models of cardiac arrhythmias’ characteristics and significance help in development of new antiarrhythmic drugs.
... Second, genetic animal models have been useful to dissect specific molecular pathways contributing to AF. We and others have previously used genetic models to detail how alterations in protein kinase activity have led to pathological alterations in Ca2 + release, 28,47 ion channel dysfunction, 48,49 and myofilament signaling in AF. 50 However, an underappreciated but growing area of study is the recognition that protein phosphatases underpin a significant degree of pathological signaling (Lenti-12C). ...
Article
BACKGROUND Atrial fibrillation (AF)—the most common sustained cardiac arrhythmia—increases thromboembolic stroke risk 5-fold. Although atrial hypocontractility contributes to stroke risk in AF, the molecular mechanisms reducing myofilament contractile function remain unknown. We tested the hypothesis that increased expression of PPP1R12C (protein phosphatase 1 regulatory subunit 12C)—the PP1 (protein phosphatase 1) regulatory subunit targeting MLC2a (atrial myosin light chain 2)—causes hypophosphorylation of MLC2a and results in atrial hypocontractility. METHODS Right atrial appendage tissues were isolated from human patients with AF versus sinus rhythm controls. Western blots, coimmunoprecipitation, and phosphorylation studies were performed to examine how the PP1c (PP1 catalytic subunit)-PPP1R12C interaction causes MLC2a dephosphorylation. In vitro studies of pharmacological MRCK (myotonic dystrophy kinase-related Cdc42-binding kinase) inhibitor (BDP5290) in atrial HL-1 cells were performed to evaluate PP1 holoenzyme activity on MLC2a. Cardiac-specific lentiviral PPP1R12C overexpression was performed in mice to evaluate atrial remodeling with atrial cell shortening assays, echocardiography, and AF inducibility with electrophysiology studies. RESULTS In human patients with AF, PPP1R12C expression was increased 2-fold versus sinus rhythm controls ( P =2.0×10 ⁻ ² ; n=12 and 12 in each group) with >40% reduction in MLC2a phosphorylation ( P =1.4×10 ⁻ ⁶ ; n=12 and 12 in each group). PPP1R12C-PP1c binding and PPP1R12C-MLC2a binding were significantly increased in AF ( P =2.9×10 ⁻² and 6.7×10 ⁻³ , respectively; n=8 and 8 in each group). In vitro studies utilizing drug BDP5290, which inhibits T560-PPP1R12C phosphorylation, demonstrated increased PPP1R12C binding with both PP1c and MLC2a and dephosphorylation of MLC2a. Mice treated with lentiviral PPP1R12C vector demonstrated a 150% increase in left atrial size versus controls ( P =5.0×10 ⁻ ⁶ ; n=12, 8, and 12), with reduced atrial strain and atrial ejection fraction. Pacing-induced AF in mice treated with lentiviral PPP1R12C vector was significantly higher than in controls ( P =1.8×10 ⁻² and 4.1×10 ⁻² , respectively; n=6, 6, and 5). CONCLUSIONS Patients with AF exhibit increased levels of PPP1R12C protein compared with controls. PPP1R12C overexpression in mice increases PP1c targeting to MLC2a and causes MLC2a dephosphorylation, which reduces atrial contractility and increases AF inducibility. These findings suggest that PP1 regulation of sarcomere function at MLC2a is a key determinant of atrial contractility in AF.
... Genetic expression of brain-heart ion channels (KCNA1 encoding voltage-gated Kv1.1 potassium channels) related both to epilepsy and AF has been demonstrated previously but were not assessed in the present study. 46,47 ...
Article
Full-text available
Objective Identification of epilepsy patients with elevated risk for atrial fibrillation (AF) is critical given the heightened morbidity and premature mortality associated with this arrhythmia. Epilepsy is a worldwide health problem affecting nearly 3.4 million people in the United States alone. The potential for increased risk for AF in patients with epilepsy is not well appreciated, despite recent evidence from a national survey of 1.4 million hospitalizations indicating that AF is the most common arrhythmia in people with epilepsy. Methods We analyzed inter‐lead heterogeneity of P‐wave morphology, a marker reflecting arrhythmogenic nonuniformities of activation/conduction in atrial tissue. The study groups consisted of 96 patients with epilepsy and 44 consecutive patients with AF in sinus rhythm before clinically indicated ablation. Individuals without cardiovascular or neurological conditions (n = 77) were also assessed. We calculated P‐wave heterogeneity (PWH) by second central moment analysis of simultaneous beats from leads II, III, and aVR (“atrial dedicated leads”) from standard 12‐lead electrocardiography (ECG) recordings from admission day to the epilepsy monitoring unit (EMU). Results Female patients composed 62.5%, 59.6%, and 57.1% of the epilepsy, AF, and control subjects, respectively. The AF cohort was older (66 ± 1.1 years) than the epilepsy group (44 ± 1.8 years, p < .001). The level of PWH was greater in the epilepsy group than in the control group (67 ± 2.6 vs. 57 ± 2.5 μV, p = .046) and reached levels observed in AF patients (67 ± 2.6 vs. 68 ± 4.9 μV, p = .99). In multiple linear regression analysis, PWH levels in individuals with epilepsy were mainly correlated with the PR interval and could be related to sympathetic tone. Epilepsy remained associated with PWH after adjustments for cardiac risk factors, age, and sex. Significance Patients with chronic epilepsy have increased PWH comparable to levels observed in patients with AF, while being ~20 years younger, suggesting an acceleration in structural change and/or cardiac electrical instability. These observations are consistent with emerging evidence of an “epileptic heart” condition.
... No reuse allowed without permission. 28,44 , ion channel dysfunction 45,46 , and myofilament signaling in AF. 47 However, an underappreciated but growing area of study is the recognition that protein phosphatases underpin a significant degree of pathologic signaling in AF. Importantly, protein phosphatase specificity is distinct from protein kinases, in that the regulatory subunits define subcellular site specificity. ...
Preprint
Full-text available
Background Atrial fibrillation (AF), the most common sustained cardiac arrhythmia, increases thromboembolic stroke risk five-fold. Although atrial hypocontractility contributes to stroke risk in AF, the molecular mechanisms reducing myofilament contractile function remain unknown. We tested the hypothesis that increased expression of PPP1R12C, the PP1 regulatory subunit targeting atrial myosin light chain 2 (MLC2a), causes hypophosphorylation of MLC2a and results in atrial hypocontractility. Methods Right atrial appendage tissues were isolated from human AF patients versus sinus rhythm (SR) controls. Western blots, co-immunoprecipitation, and phosphorylation studies were performed to examine how the PP1c-PPP1R12C interaction causes MLC2a de-phosphorylation. In vitro studies of pharmacologic MRCK inhibitor (BDP5290) in atrial HL-1 cells were performed to evaluate PP1 holoenzyme activity on MLC2a. Cardiac-specific lentiviral PPP1R12C overexpression was performed in mice to evaluate atrial remodeling with atrial cell shortening assays, echocardiography, and AF inducibility with EP studies. Results In human patients with AF, PPP1R12C expression was increased two-fold versus SR controls ( P =2.0×10 ⁻² , n=12,12 in each group) with > 40% reduction in MLC2a phosphorylation ( P =1.4×10 ⁻⁶ , n=12,12 in each group). PPP1R12C-PP1c binding and PPP1R12C-MLC2a binding were significantly increased in AF ( P =2.9×10 ⁻² and 6.7×10 ⁻³ respectively, n=8,8 in each group). In vitro studies utilizing drug BDP5290, which inhibits T560-PPP1R12C phosphorylation, demonstrated increased PPP1R12C binding with both PP1c and MLC2a, and dephosphorylation of MLC2a. Lenti-12C mice demonstrated a 150% increase in LA size versus controls ( P =5.0×10 ⁻⁶ , n=12,8,12), with reduced atrial strain and atrial ejection fraction. Pacing-induced AF in Lenti-12C mice was significantly higher than controls ( P =1.8×10 ⁻² and 4.1×10 ⁻² respectively, n= 6,6,5). Conclusions AF patients exhibit increased levels of PPP1R12C protein compared to controls. PPP1R12C overexpression in mice increases PP1c targeting to MLC2a and causes MLC2a dephosphorylation, which reduces atrial contractility and increases AF inducibility. These findings suggest that PP1 regulation of sarcomere function at MLC2a is a key determinant of atrial contractility in AF.
... Kv1.1 subunit can inhibit neuronal membrane excitability and regulate the discharge characteristics of action potential [97]. Despite the low expression, Kv1.1 subunits were actually found in cardiomyocytes, where they control the morphology of action potential and promote arrhythmia susceptibility [98,99]. Neuron-specific Kcna1 deletion could partially reproduce many significant phenotypes observed in Kcna1 global KO mice, including epilepsy, premature death and cardiopulmonary dysfunction, which indicated that these defects are mainly driven by the nervous system. ...
Article
Full-text available
Epilepsy is one of the most common neurological disorders, and sudden unexpected death in epilepsy (SUDEP) is the most severe outcome of refractory epilepsy. Arrhythmia is one of the heterogeneous factors in the pathophysiological mechanism of SUDEP with a high incidence in patients with refractory epilepsy, increasing the risk of premature death. The gene co-expressed in the brain and heart is supposed to be the genetic basis between epilepsy and arrhythmia, among which the gene encoding ion channel contributes to the prevalence of “cardiocerebral channelopathy” theory. Nevertheless, this theory could only explain the molecular mechanism of comorbid arrhythmia in part of patients with epilepsy (PWE). Therefore, we summarized the mutant genes that can induce comorbidity of epilepsy and arrhythmia and the possible corresponding treatments. These variants involved the genes encoding sodium, potassium, calcium and HCN channels, as well as some non-ion channel coding genes such as CHD4, PKP2, FHF1, GNB5, and mitochondrial genes. The relationship between genotype and clinical phenotype was not simple linear. Indeed, genes co-expressed in the brain and heart could independently induce epilepsy and/or arrhythmia. Mutant genes in brain could affect cardiac rhythm through central or peripheral regulation, while in the heart it could also affect cerebral electrical activity by changing the hemodynamics or internal environment. Analysis of mutations in comorbidity of epilepsy and arrhythmia could refine and expand the theory of “cardiocerebral channelopathy” and provide new insights for risk stratification of premature death and corresponding precision therapy in PWE.
Chapter
Atrial fibrillation is triggered when an abnormal electrical impulse encounters a substrate that allows initiation of re-entry. The arrhythmia has a strong tendency to become persistent due to structural and electrical remodeling. Currently available antiarrhythmic drugs are not sufficiently effective and are burdened by cardiac and extracardiac side effects that may offset their therapeutic benefits. Detailed knowledge about the mechanisms leading to generation and maintenance of arrhythmias may lead to the discovery of new targets for pharmacological interventions. These could include atria-selective ion channels (e.g., Nav1.5 (INa,), Kv1.5 (IKur), Kir3.1/3.4 (IK,ACh), TASK-1 (ITASK-1), and SK (IK,Ca) channels, pathology-selective ion channels (constitutively active Kir3.x, TRP channels), ischemia-uncoupled gap junctions, proteins related to malfunctioning intracellular Ca2+ homeostasis (e.g., “leaky” ryanodine receptors, overactive Na+,Ca2+ exchanger), or risk factors for arrhythmias (“upstream” therapies). In this chapter, ion channels as drug targets for atrial fibrillation will be discussed in the context of the remodeled atria.
Article
Full-text available
Cardiac rhythm regulated by micro-macroscopic structures of heart. Pacemaker abnormalities or disruptions in electrical conduction, lead to arrhythmic disorders may be benign, typical, threatening, ultimately fatal, occurs in clinical practice, patients on digitalis, anaesthesia or acute myocardial infarction. Both traditional and genetic animal models are: In-vitro: Isolated ventricular Myocytes, Guinea pig papillary muscles, Patch-Clamp Experiments, Porcine Atrial Myocytes, Guinea pig ventricular myocytes, Guinea pig papillary muscle: action potential and refractory period, Langendorff technique, Arrhythmia by acetylcholine or potassium. Acquired arrhythmia disorders: Transverse Aortic Constriction, Myocardial Ischemia, Complete Heart Block and AV Node Ablation, Chronic Tachypacing, Inflammation, Metabolic and Drug-Induced Arrhythmia. In-Vivo: Chemically induced arrhythmia: Aconitine antagonism, Digoxin-induced arrhythmia, Strophanthin/ouabain-induced arrhythmia, Adrenaline-induced arrhythmia, and Calcium-induced arrhythmia. Electrically induced arrhythmia: Ventricular fibrillation electrical threshold, Arrhythmia through programmed electrical stimulation, sudden coronary death in dogs, Exercise ventricular fibrillation. Genetic Arrhythmia: Channelopathies, Calcium Release Deficiency Syndrome, Long QT Syndrome, Short QT Syndrome, Brugada Syndrome. Genetic with Structural Heart Disease: Arrhythmogenic Right Ventricular Cardiomyopathy/Dysplasia, Dilated Cardiomyopathy, Hypertrophic Cardiomyopathy, Atrial Fibrillation, Sick Sinus Syndrome, Atrioventricular Block, Preexcitation Syndrome. Arrhythmia in Pluripotent Stem Cell Cardiomyocytes. Conclusion: Both traditional and genetic, experimental models of cardiac arrhythmias’ characteristics and significance help in development of new antiarrhythmic drugs.
Article
Cardiac arrhythmias are a significant cause of morbidity and mortality worldwide, accounting for 10% to 15% of all deaths. Although most arrhythmias are due to acquired heart disease, inherited channelopathies and cardiomyopathies disproportionately affect children and young adults. Arrhythmogenesis is complex, involving anatomic structure, ion channels and regulatory proteins, and the interplay between cells in the conduction system, cardiomyocytes, fibroblasts, and the immune system. Animal models of arrhythmia are powerful tools for studying not only molecular and cellular mechanism of arrhythmogenesis but also more complex mechanisms at the whole heart level, and for testing therapeutic interventions. This review summarizes basic and clinical arrhythmia mechanisms followed by an in-depth review of published animal models of genetic and acquired arrhythmia disorders.
Article
Full-text available
The sustained component of the K+ outward current in human atrial myocytes is believed to be due to the slowly inactivating ultra-rapid potassium current I Kur and not to the fast inactivating transient outward current I to. Here we provide evidence for contribution of I to to this late current due to the effects of dipeptidyl peptidase-like protein (DPP) 10 (DPP10a) interacting with Kv4.3 channels. We studied the late current component of I to in human atrial myocytes and CHO cells co-expressing Kv4.3 or Kv4.3/KChIP2 (control) and DPP proteins using voltage-clamp technique and a pharmacological approach. A voltage dependent and slowly inactivating late current (43 % of peak amplitude) could be observed in atrial myocytes. We found a similar current in CHO cells expressing Kv4.3/KChIP2 + DPP10a, but not in cells co-expressing Kv4.3 + DPP or Kv4.3/KChIP2 + DPP6-S. Assuming that DPP10a influences atrial I to, we detected DPP10 expression of three alternatively spliced mRNAs, DPP10 protein and colocalization of Kv4.3 and DPP10 proteins in human atrial myocytes. DPP10a did not affect properties of expressed Kv1.5 excluding a contribution to the sustained I Kur in atrial cells. To test for the contribution of Kv4-based I to on sustained K+ outward currents in human atrial myocytes, we used 4-AP to block I Kur, in combination with Heteropoda toxin 2 to block Kv4 channels. We could clearly separate an I to fraction of about 19 % contributing to the late current in atrial myocytes. Thus, the interaction of DPP10a, expressed in human atrium, with Kv4.3 channels generates a sustained current component of I to, which may affect late repolarization phase of atrial action potentials.
Article
Full-text available
Electrical, structural, and Ca(2+)-handling remodeling contribute to the perpetuation/progression of atrial fibrillation (AF). Recent evidence has suggested a role for spontaneous sarcoplasmic reticulum Ca(2+)-release events in long-standing persistent AF, but the occurrence and mechanisms of sarcoplasmic reticulum Ca(2+)-release events in paroxysmal AF (pAF) are unknown. Right-atrial appendages from control sinus rhythm patients or patients with pAF (last episode a median of 10-20 days preoperatively) were analyzed with simultaneous measurements of [Ca(2+)]i (fluo-3-acetoxymethyl ester) and membrane currents/action potentials (patch-clamp) in isolated atrial cardiomyocytes, and Western blot. Action potential duration, L-type Ca(2+) current, and Na(+)/Ca(2+)-exchange current were unaltered in pAF, indicating the absence of AF-induced electrical remodeling. In contrast, there were increases in SR Ca(2+) leak and incidence of delayed after-depolarizations in pAF. Ca(2+)-transient amplitude and sarcoplasmic reticulum Ca(2+) load (caffeine-induced Ca(2+)-transient amplitude, integrated Na(+)/Ca(2+)-exchange current) were larger in pAF. Ca(2+)-transient decay was faster in pAF, but the decay of caffeine-induced Ca(2+) transients was unaltered, suggesting increased SERCA2a function. In agreement, phosphorylation (inactivation) of the SERCA2a-inhibitor protein phospholamban was increased in pAF. Ryanodine receptor fractional phosphorylation was unaltered in pAF, whereas ryanodine receptor expression and single-channel open probability were increased. A novel computational model of the human atrial cardiomyocyte indicated that both ryanodine receptor dysregulation and enhanced SERCA2a activity promote increased sarcoplasmic reticulum Ca(2+) leak and sarcoplasmic reticulum Ca(2+)-release events, causing delayed after-depolarizations/triggered activity in pAF. Increased diastolic sarcoplasmic reticulum Ca(2+) leak and related delayed after-depolarizations/triggered activity promote cellular arrhythmogenesis in pAF patients. Biochemical, functional, and modeling studies point to a combination of increased sarcoplasmic reticulum Ca(2+) load related to phospholamban hyperphosphorylation and ryanodine receptor dysregulation as underlying mechanisms.
Article
Full-text available
Significance The heart is continuously subjected to mechanical forces. The atria in particular are susceptible to changes in the mechanical environment due to their unique position as “pressure sensors.” Here, we show that increased shear stress induces the recruitment of potassium channels from intracellular storage pools to the plasma membrane, via signaling pathways that link the extracellular matrix to the cytoskeleton. This process is altered in myocytes experiencing chronically increased mechanical stress. The incorporation of channels into the membrane causes changes in the electrical activity of the myocyte and may be an important way for cells to adapt to increased mechanical forces.
Article
Full-text available
Aims: Transverse tubules (TTs) provide the basic subcellular structures that facilitate excitation-contraction (EC) coupling, the essential process that underlies normal cardiac contractility. Previous studies have shown that TTs develop within the first few weeks of life in mammals but the molecular determinants of this development have remained elusive. This study aims to elucidate the role of junctophilin-2 (JPH2), a junctional membrane complex protein, in the maturation of TTs in cardiomyocytes. Methods and results: Using a novel cardiac-specific short-hairpin-RNA-mediated JPH2 knockdown mouse model (Mus musculus; αMHC-shJPH2), we assessed the effects of the loss of JPH2 on the maturation of the ventricular TT structure. Between embryonic day (E) 10.5 and postnatal day (P) 10, JPH2 mRNA and protein levels were reduced by >70% in αMHC-shJPH2 mice. At P8 and P10, knockdown of JPH2 significantly inhibited the maturation of TTs, while expression levels of other genes implicated in TT development remained mostly unchanged. At the same time, intracellular Ca(2+) handling was disrupted in ventricular myocytes from αMHC- shJPH2 mice, which developed heart failure by P10 marked by reduced ejection fraction, ventricular dilation, and premature death. In contrast, JPH2 transgenic mice exhibited accelerated TT maturation by P8. Conclusion: Our findings suggest that JPH2 is necessary for TT maturation during postnatal cardiac development in mice. In particular, JPH2 may be critical in anchoring the invaginating sarcolemma to the sarcoplasmic reticulum, thereby enabling the maturation of the TT network.
Article
Objective: To investigate changes in human atrial single cell functional electrophysiological properties associated with chronic atrial fibrillation (AF), and the contribution to these of accompanying ion current changes. Methods: The whole cell patch clamp technique was used to record action potentials, the effective refractory period (ERP) and ion currents, in the absence and presence of drugs, in enzymatically isolated myocytes from 11 patients with chronic (<6 months) AF and 39 patients in sinus rhythm. Results: Stimulation at high rates (up to 600 beats/min) markedly shortened late repolarisation and the ERP in cells from patients in sinus rhythm, and depolarised the maximum diastolic potential (MDP). Chronic AF was associated with a reduction in the ERP at physiological rate (from 203±16 to 104±15 ms, P<0.05), and marked attenuation in rate effects on the ERP and repolarisation. The abbreviated terminal phase of repolarisation prevented fast rate-induced depolarisation of the MDP in cells from patients with AF. The density of L-type Ca2+ (ICaL) and transient outward K+ (ITO) currents was significantly reduced in cells from patients with AF (by 60–65%), whilst the inward rectifier K+ current (IK1) was increased, and the sustained outward current (IKSUS) was unaltered. Superfusion of cells from patients in sinus rhythm with nifedipine (10 micromol/l) moderately shortened repolarisation, but had no effect on the ERP (228±12 vs. 225±11 ms). 4-Aminopyridine (2 mmol/l) markedly prolonged repolarisation and the ERP (by 35%, P<0.05). However, the combination of these drugs had no effect on late repolarisation or refractoriness. Conclusion: Chronic AF in humans is associated with attenuation in adaptation of the atrial single cell ERP and MDP to fast rates, which may not be explained fully by accompanying changes in ICaL and ITO.
Article
The two most commonly used methods to analyze data from real-time, quantitative PCR experiments are absolute quantification and relative quantification. Absolute quantification determines the input copy number, usually by relating the PCR signal to a standard curve. Relative quantification relates the PCR signal of the target transcript in a treatment group to that of another sample such as an untreated control. The 2(-DeltaDeltaCr) method is a convenient way to analyze the relative changes in gene expression from real-time quantitative PCR experiments. The purpose of this report is to present the derivation, assumptions, and applications of the 2(-DeltaDeltaCr) method. In addition, we present the derivation and applications of two variations of the 2(-DeltaDeltaCr) method that may be useful in the analysis of real-time, quantitative PCR data. (C) 2001 Elsevier science.
Article
OBJECTIVES Our purpose was to determine whether patients with persistent atrial fibrillation (AF) and patients with paroxysmal AF show alterations in potassium channel expression.BACKGROUND Persistent AF is associated with a sustained shortening of the atrial action potential duration and atrial refractory period. Underlying molecular changes have not been studied in humans. We investigated whether a changed gene expression of specific potassium channels is associated with these changes in patients with persistent AF and in patients with paroxysmal AF.METHODS Right atrial appendages were obtained from 8 patients with paroxysmal AF, 10 with persistent AF and 18 matched controls in sinus rhythm. All controls underwent coronary artery bypass surgery, whereas most AF patients underwent Cox’s MAZE surgery (atrial arrhythmia surgery to cure AF) (n = 12). All patients had normal left ventricular function. mRNA (ribonucleic acid) levels were measured by semiquantitative polymerase chain reaction and protein content by Western blotting.RESULTSmRNA levels of transient outward channel (Kv4.3), acetylcholine-dependent potassium channel (Kir3.4) and ATP-dependent potassium channel (Kir6.2) were reduced in patients with persistent AF (−35%, −47% and −36%, respectively, p < 0.05), whereas only Kv4.3 mRNA level was reduced in patients with paroxysmal AF (−29%, p = 0.03). No changes were found for Kv1.5 and HERG mRNA levels in either group. Protein levels of Kv4.3, Kv1.5 and Kir3.1 were reduced both in patients with persistent AF (−39%, −84% and −47%, respectively, p < 0.05) and in those with paroxysmal AF (−57%, −64%, and −40%, respectively, p < 0.05).CONCLUSIONS Persistent AF is accompanied by reductions in mRNA and protein levels of several potassium channels. In patients with paroxysmal AF these reductions were observed predominantly at the protein level and not at the mRNA level, suggesting a post-transcriptional regulation.
Article
Key points Voltage‐gated Kv1.1 potassium channels cluster at juxtaparanodes of myelinated axons in the vagus nerve, which provides parasympathetic innervation to the heart. Kcna1 knockout mice lacking Kv1.1 channels exhibit frequent atrioventricular cardiac conduction blocks that are abolished by atropine, suggestive of a vagal mechanism. Electrophysiological analysis of single myelinated axons from wild‐type and Kv1.1‐deficient mouse vagus nerves revealed that the absence of Kv1.1 channels rendered large myelinated vagal axons far more susceptible to spontaneous ectopic firing in the presence of 4‐aminopyridine. KCNQ2 potassium channels are present within vagal nodes of Ranvier and their activation with flupirtine rescued single axon hyperexcitability mediated by juxtaparanodal Kv1.1‐deficiency. These results demonstrate a functional synergy between nodal and extranodal K ⁺ channels and implicate KCNQ channels as potential targets for Kv1‐related peripheral nerve hyperexcitability. Abstract Kv1.1 channels cluster at juxtaparanodes of myelinated axons in the vagus nerve, the primary conduit for parasympathetic innervation of the heart. Kcna1 ‐null mice lacking these channels exhibit neurocardiac dysfunction manifested by atropine‐sensitive atrioventricular conduction blocks and bradycardia that may culminate in sudden death. To evaluate whether loss of Kv1.1 channels alters electrogenic properties within the nerve, we compared the intrinsic excitability of single myelinated A‐ and Aδ‐axons from excised cervical vagus nerves of young adult Kcna1 ‐null mice and age‐matched, wild‐type littermate controls. Although action potential shapes and relative refractory periods varied little between genotypes, Kv1.1‐deficient large myelinated A‐axons showed a fivefold increase in susceptibility to 4‐aminopyridine (4‐AP)‐induced spontaneous ectopic firing. Since the repolarizing currents of juxtaparanodal Kv1 channels and nodal KCNQ potassium channels both act to dampen repetitive activity, we examined whether augmenting nodal KCNQ activation could compensate for Kv1.1 loss and reverse the spontaneous hyperexcitability in Kv1.1‐deficient A‐axons. Application of the selective KCNQ opener flupirtine raised A‐axon firing threshold while profoundly suppressing 4‐AP‐induced spontaneous firing, demonstrating a functional synergy between the two compartments. We conclude that juxtaparanodal Kv1.1‐deficiency causes intrinsic hyperexcitability in large myelinated axons in vagus nerve which could contribute to autonomic dysfunction in Kcna1 ‐null mice, and that KCNQ openers reveal a transcompartmental synergy between Kv1 and KCNQ channels in regulating axonal excitability.