Access to this full-text is provided by Wiley.
Content available from Cardiovascular Therapeutics
This content is subject to copyright. Terms and conditions apply.
Review Article
Research Progress on Natural Products’Therapeutic Effects on
Atrial Fibrillation by Regulating Ion Channels
Jinshan He ,
1
Sicong Li ,
2
Yumeng Ding ,
3
Yujia Tong ,
4
and Xuebin Li
1
1
Cardiovascular Department, Peking University People’s Hospital, Beijing, China
2
School of Pharmacy, Peking University Health Science Centre, Beijing, China
3
School of Life Sciences, Shanxi Normal University, Shanxi, China
4
Institute of Medical Information, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
Correspondence should be addressed to Xuebin Li; docxuebin.li@vip.sina.com
Received 26 August 2021; Revised 28 January 2022; Accepted 3 March 2022; Published 22 March 2022
Academic Editor: Simona Saponara
Copyright © 2022 Jinshan He et al. This is an open access article distributed under the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Antiarrhythmic drugs (AADs) have a therapeutic effect on atrial fibrillation (AF) by regulating the function of ion channels.
However, several adverse effects and high recurrence rates after drug withdrawal seriously affect patients’medication
compliance and clinical prognosis. Thus, safer and more effective drugs are urgently needed. Active components extracted
from natural products are potential choices for AF therapy. Natural products like Panax notoginseng (Burk.) F.H. Chen,
Sophora flavescens Ait., Stephania tetrandra S. Moore., Pueraria lobata (Willd.) Ohwi var. thomsonii (Benth.) Vaniot der
Maesen., and Coptis chinensis Franch. have a long history in the treatment of arrhythmia, myocardial infarction, stroke, and
heart failure in China. Based on the classification of chemical structures, this article discussed the natural product components’
therapeutic effects on atrial fibrillation by regulating ion channels, connexins, and expression of related genes, in order to
provide a reference for development of therapeutic drugs for atrial fibrillation.
1. Introduction
Atrial fibrillation (AF) is associated with a higher risk of
stroke, heart failure with reduced ejection fraction, cardiomy-
opathy, acute coronary syndrome, and impaired quality of life.
As shown in Figure 1, the pathophysiological changes of atrial
fibrillation include oxidative stress, atrial structure, electrical
remodeling, autonomic nerve dysfunction, metabolic abnor-
malities, ectopic activation, and reentry. AF treatment mainly
includes rate and rhythm control therapy, anticoagulation,
and left atrial appendage closure. Restoration and maintaining
sinus rhythm have shown superiority in improving survival,
quality of life, and ventricular function and reducing heart fail-
ure hospitalization. British National Institute for Health and
Care Excellence (NICE) guideline [1], 2019 American Heart
Association (AHA)/American College of Cardiology (ACC)/
Heart Rhythm Society (HRS) AF guideline [2], and 2020 the
European Association of Cardio-Thoracic Surgery (EACTS)
[3] all recommended catheter ablation (mainly including
radiofrequency ablation and cryoballoon ablation) for sinus
rhythm recovery in patients with atrial fibrillation. However,
after primary catheter ablation, the recurrence rate of AF/AF
is high, especially in patients with severe structural cardiac
remodeling, chronic kidney disease, and hyperthyroidism.
Some patients have to receive more than one catheter ablation
or be followed with electric cardioversion [4].
AADs have a therapeutic effect on atrial fibrillation by
regulating potassium, calcium, sodium channels, or β1
receptors. However, adverse reactions can seriously affect
the prognosis of patients with atrial fibrillation. Propafenone,
sotalol, and ibutilide may give rise to severe ventricular
arrhythmia. Amiodarone may cause interstitial lung disease,
thyroid dysfunction, and nonalcoholic fatty liver disease.
Natural products have unique advantages in antiarrhythmia,
with few side effects and rarely inducing other arrhythmias.
In recent years, it has become a hotspot to search for antiar-
rhythmia active ingredients from natural products. In this
article, natural products with effects on more than two kinds
of ion channels were included. By taking chemical structure
as classification standard, natural products’antiarrhythmic
Hindawi
Cardiovascular erapeutics
Volume 2022, Article ID 4559809, 23 pages
https://doi.org/10.1155/2022/4559809
effects on ion channels and target genes were discussed in
detail to lay a foundation for the follow-up research and
development of antiarrhythmic drugs.
2. Ion Channels and Connexins in the
Pathogenesis of Atrial Fibrillation
As signal detectors, relayers, and amplifiers, ion channels
regulate signal transduction and ion transport across cell
membranes [5]. The abnormal function of ion channels
increases vulnerablity and sustainability of AF [6]. Recur-
rence of atrial fibrillation can lead to a shorter effective
refractory period of atrial myocytes, increased dispersion,
and decreased or disappearance of frequency adaptability,
thus promoting the deterioration of paroxysmal atrial fibril-
lation into persistent atrial fibrillation. Moreover, ion chan-
nel coding genes related to atrial fibrillation have also been
discovered. In patients with atrial fibrillation, decreased
expression of L-type Ca 2 + channel, ryanodine receptor
(RyR2), potassium voltage-gated channel subfamily A
member 5 (KCNA5), sarcoplasmic reticular Ca2+-ATPase
(SERCA2), the beta-subunit MinK (KCNE1) and MIRP2
(KCNE3) [7], and increased expression of
hyperpolarization-activated cation channel two associated
with the pacemaker current I (f) (HCN2) were observed
[8]. The left atrial diameter was negatively correlated with
the expression of RyR2 and KChIP2 [7].
2.1. Potassium Channel. Four major kinds of potassium
channels have been identified in cardiovascular myocytes,
including inwardly rectifying (Kir) K+ channels, calcium-
activated potassium channel (KCa), voltage-gated (VK),
and two-pore domain potassium channels (K2P) [5]. (Kir)
current (IK1) and acetylcholine-activated potassium current
(IK, Ach) are involved in AF drivers formation (rotors and
focal impulses) [9–11]. The increase of vagus nerve tension
can enhance IKAch to shorten APD and stabilize the rotor
related to reentry [12]. KCa channels can be divided into
the big, small, and intermediate-conductance K+ channels
[13]. KCa channels are also related to atrial structural and
electrical remodeling in atrial fibrillation [14]. Transient out-
ward current (Ito), similarly to the ultrarapid current (IKur),
contributes to early repolarization [15]. By starting
membrane depolarization, VK allows K+ efflux and regu-
lates cardiac action potential duration. K2P- channels
conduct outward K+ currents and modulate action potential
repolarization [16].
2.2. Calcium Channel. L-type calcium current is the primary
inward current in the action potential plateau phase, while
T-type calcium current depolarizes current in phase 0 of
action potential duration [17]. Generated and conducted
from the sinoatrial node and atrioventricular node, L-type
Ca2+ current was not only the primary inward current in
atrial and ventricular action potential 2 phases [18]. Depo-
larization of cardiomyocytes can open the L-type calcium
channel and the influx of Ca2+ and then trigger the release
of Ca2+ from the sarcoplasmic reticulum. The process above
is essential in the excitation-contraction coupling of cardio-
myocytes [19].
Abnormal intracellular calcium (Ca2+) handling can
trigger delayed after depolarization (DADs) and thus
Atrial
brillation
Rhythm control
Rate control
Heart failure
myocardial infarction stroke
Anticoagulation
Le atrial appendage closure
(LAAC)
Electrical
remodeling
Structural
remodeling
Ectopic
focus and
reentry
Oxidative
stress
Abnormal
metabolism
Autonomic
nerve
dysfunction
ROS
Figure 1: Pathophysiological changes, treatment, and complications of atrial fibrillation.
2 Cardiovascular Therapeutics
increase atrial ectopic activity. Intracellular calcium- (Ca2
+-) calmodulin- (CaM-) calmodulin kinase (CaMK II) signal
transduction pathway plays a central role in the regulation of
intracellular calcium. Increased spontaneous sarcoplasmic
reticulum (SR) Ca2+ release leads to ryanodine receptor
(RyR2) dysregulation and Ca2+/calmodulin-dependent
protein kinase II (CaMKII) hyperactivity. Exciting βadre-
noceptors can enhance RyR2 receptor phosphorylation
and promote ectopic activation associated with delayed
depolarization [20]
2.3. Sodium Channel. The voltage-gated sodium channels
contribute to the initiation and conduction of action poten-
tial [21, 22]. Prolonging sodium influx in the plateau phase
can lead to early afterdepolarizations and ventricular tachy-
cardia [23]. The sequential activation and inactivation of
sodium channels prevent proarrhythmic events [24]. In
recent years, late sodium current has been related to atrial
fibrillation [25]. It promotes the occurrence of AF by
increasing the dispersion of repolarization and leads to
intracellular calcium overload.
2.4. Connexin. Cardiac connexins contribute to gap junc-
tions intercellular electrical and molecular signaling commu-
nication [26]. There are three different connexins in the
human heart, including Cx40, Cx43, and Cx45 [27, 28].
Connexin 45 is mainly expressed in sinus node (SA) and
atrioventricular node (AV), while connexin 43 and connexin
40 are both expressed in atrial muscle [29]. Cx40 promoter
polymorphisms that inhibit the expression of Cx40 are asso-
ciated with the early onset of AF [30]. Somatic and germline
mutations within the coding regions of the human Cx40
gene (GJA5) are also related to a higher risk of AF [31].
Atrial fibrillation leads to less connexin protein, less intercel-
lular electrical coupling, changes in the electrical conductiv-
ity of the myocardium, the conduction velocity, and an
aggravated degree of the auriculoventricular block.
2.5. Hyperpolarization-Activated Cyclic Nucleotide-Gated
Cation Channel (HCN). HCN2 and HCN4 are channels
responsible for cardiac hyperpolarization-activated cation
current (or “funny current”, If) [32, 33]. Their function
decreases in the sinoatrial node and increases in the atrial
and pulmonary vein in atrial fibrillation patients [34].
Activated by intracellular cAMP and membrane hyperpolar-
ization, cardiac hyperpolarization-activated cation current
contributes to diastolic depolarization of pacemaker cells [35].
3. Natural Products with Bioactivity in AF
3.1. Saponin. Saponins are glycosides with triterpenes or
helical steranes as aglycones. Saponins are the main practical
components of Panax notoginseng (Burkill) F. H. Chen ex C.
H., Radix Polygalae, Platycodon grandiflorus, and Radix
Bupleuri [36].
3.1.1. Panax notoginseng Saponins (PNS). Extracted from the
roots of Panax notoginseng (Burk.) F.H. Chen, PNS has
antiarrhythmia, antiplatelet [37], antishock [38], antioxida-
tion [39], sedative [40], and antitumor [41] effects. Panax
notoginseng saponins have been approved in the treatment
of central retinal vein occlusion, sequelae of cerebrovascular
disease, enophthalmos, and anterior chamber hemorrhage in
China. By regulating potassium and calcium ion channels,
PNS reduced the automaticity of cardiomyocytes, slowed
down cardiac electrical conduction, prolonged action poten-
tial duration (APD) and effective refractory period (ERP),
and prevented reentry agitation [42].
(1) Potassium Channels. PNS (150 mg/kg intraperitoneal
injected once) significantly inhibited myocardial cell apopto-
sis induced by isoproterenol in atrial fibrillation model mice
[43]. PNS significantly downregulated the expression of
miR-499 in atrial tissues compared with the control group
(P<0:05). Small conductance calcium-activated potassium
channel 3 (SK3) plays an essential role in the development
of atrial fibrillation. Knocking out the SK3 gene could trigger
atrial fibrillation. By downregulating, the expression of
potassium calcium-activated channel subfamily N member
3(KCNN3) and SK3 and microRNA-499 (miR-499) affected
the activity of the SK3 pathway and then triggered the occur-
rence of atrial fibrillation.
(2) Calcium Channel. CACNA1C gene encoded the L-type
Ca2+ channel α1 subunit (Cav1.2) [44]. PNS (0.1, 0.6, 1,
and 4 g/L) [45] inhibited L-type calcium channels (Cav1.2)
and T-type calcium channels (Cav3.1) in Xenopus laevis
oocytes in vitro. MicroRNA29 (miR29) targeted extracellular
matrix proteins. It played an important role in the atrial
fibrotic remodeling of AF and chronic heart failure patients
[46]. By regulating the expression of the ATPase sarcoplas-
mic/endoplasmic reticulum Ca2+ receptor 2(ATP2A2) gene,
miR-328 affected the intracellular Ca2+ concentration and
the expression of calcineurin [47]. CACNA1C and CACNB1
that respectively encoded α1c- and β1 subunits of cardiac L-
type Ca2+ channel were potential targets of miR-328 [48].
PNS [44] (150 mg/kg intraperitoneal injected once) signifi-
cantly upregulated the expression of miR-29b and miR-328
(P<0:05), thereby inhibiting isoproterenine-induced atrial
fibrillation. In addition, PNS reduced the release of calcium
in the sarcoplasmic reticulum, thus improving the apoptosis
of myocardial cells caused by calcium overload [49].
In addition to regulating ion channels, the therapeutic
effects of PNS on atrial fibrillation were also associated with
anti-inflammation, antifibrosis, and antioxidative stress
effects. After seven days of intraperitoneal injecting PNS
(100 μg/g), atrial fibrillation induced by ACh-CaCl2 mixed
solution was significantly inhibited with effective refractory
period prolonged, and the duration of atrial fibrillation
shortened [50]. By activating the PI3K-AKT signaling path-
way, the infiltration of inflammatory cells into cardiomyo-
cytes and blood vessels and the deposition of collagen
fibers around blood vessels were inhibited, with myocardial
fibrosis improved in rats with atrial fibrillation [51].
In clinical trials, the therapeutic effect of PNS on atrial
fibrillation has also been confirmed. Thirty-five patients with
atrial fibrillation in the treatment group were given PNS
3Cardiovascular Therapeutics
(orally taking 100 mg, tid, for six months) combined with
amiodarone (orally taking 200 mg, tid, for the first week,
200 mg bid for the second week, and continued with
200 mg once a day until six months), and 35 patients in
the control group were given amiodarone (the same dosage
as above, for six months) [52]. The AF recurrence rate in
the treatment group was significantly lower than that of
the control group (14.29% vs. 40%, P<0:05). Left atrial
diameter and ankyrin repeat expression in the treatment
group were lower than those in the control group after six
months of treatment. These results suggested that PNS
inhibited atrial remodeling and ectopic pacing.
3.2. Alkaloids. Alkaloids are alkaline organic compounds
containing nitrogen. They have alkaline properties and are
widely distributed in advanced plants, especially in dicotyle-
donous plants.
3.2.1. Berberine. Extracted from Coptis chinensis Franch. of
Ranunculaceae and Mahonia fortunei (Lindl.) Fedde of ber-
beris [53], berberine is a kind of isoquinoline alkaloid with
antiarrhythmia [54], anti-inflammatory [55], antibacterial
[56], hypoglycemia [57], vasodilation [58], and antitumor
effects [59]. In terms of antiarrhythmia, berberine can
prolong the action potential duration, and effective refrac-
tory period of myocardial cells [60] inhibits the occurrence
of atrioventricular reentrant tachycardia by regulating potas-
sium, calcium ion channels, and hyperpolarization-activated
cyclic nucleotide-gated cation channel [61].
(1) Potassium Channel. Berberine could inhibit the rapid and
slow activation of delayed rectifier outward potassium cur-
rent in cardiomyocytes [62]. Berberine (3-100 μmol/L)
inhibited cromakalim-induced outward currents in isolated
guinea-pig ventricular myocytes and ATP-sensitive K+
(KATP) channels [63]. Berberine (100 μmol/L) had a block-
ing effect on inward rectifier potassium current (IK1) and
outward delayed rectifier potassium current (IK) expressed
in Xenopus oocytes. In vivo, berberine (10 and 20 mg/kg)
inhibited the expression of KCNH2 in rat myocardium
(P<0:05) [64, 65].
(2) Calcium Channel. In vitro, berberine (10 and 30 μmol/L)
could not only inhibit the L-type and T-type calcium chan-
nel of guinea pig ventricular myocytes and inhibit extracellu-
lar Ca2+ influx but also reduce the delayed depolarizing
induced by calcium overload [66, 67]. CPU86017 was a ber-
berine derivative that could relieve heart failure by inhibiting
calcium leakage, downregulating phosphatase, and exerting
antioxidant activity [68]. Moreover, CPU86017 led to a
regression of the transmural dispersion of repolarization
and inhibition of RyR2 and SERCA2 [69].
(3) Hyperpolarization-Activated Cyclic Nucleotide-Gated
Cation Channel (HCN). Berberine (1-300 μmol/L) inhibited
the current of hyperpolarization-activated cyclic
nucleotide-gated 4 (hHCN4) channel expressed in Xenopus
laevis oocytes. Berberine decreased the rate of pacemaker
firing and diastolic depolarization and changed the potential
action parameters [70].
In clinical trials, Zheng et al. [71] conducted a retrospec-
tive cohort study of 88 patients with paroxysmal atrial fibril-
lation. Forty-five patients orally took berberine (the average
dose of 1.3 g/day for one year), and 43 patients orally took
amiodarone (0.2 g tid for the first week, 0.2 g bid for the sec-
ond one, and 0.2 g for the following weeks, lasted for one
year). There was no significant difference in the conversion
rate and echocardiographic parameters between the berber-
ine and amiodarone groups after 12 months of treatment.
Echocardiographic parameters showed that the E/A ratio
and left atrial diameter were significantly improved after 6
and 12 months of berberine treatment. However, in the ami-
odarone group, only E/A ratio got considerably enhanced.
3.2.2. Tetrandrine (Tet). Extracted from rhizomes of Tricho-
santhes Merr. Chun. of tetrandrine and roots of Stephania
discolor, Stephania tetrandra, and Aristolochia heterophylla,
tetrandrine is a kind of dibenzyl isoquinoline alkaloid [72]
with antiarrhythmia [73], antihypertensive [74], anti-
inflammatory [75], and antitumor [74] effects [76]. In terms
of antiarrhythmia, by inhibiting calcium, potassium, and
sodium channels, tetrandrine can slow down the heart rate,
inhibit atrioventricular conduction, and prolong the effective
refractory period of cardiomyocytes [77].
(1) Potassium Channel. Tetrandrine dosage-dependently
inhibited delayed rectifier potassium current. The maximum
effective concentration is 3×10
−5mol/L [18]. Tetrandrine
had a bidirectional regulation effect on calcium-activated
potassium channels. In vitro, tetrandrine (7.5 and
15 μmol/L) increased the opening frequency and prolonged
the opening time of calcium-activated potassium channels
in rabbit cardiomyocytes. However, at the concentration
of 30 μmol/L, it significantly reduced the opening
frequency and shortened the opening time of calcium-
activated potassium channels.
(2) Calcium Channel. Tetrandrine could inhibit both L-type
and T-type calcium channels in cardiomyocytes [78, 79].
Tetrandrine (6 μmol/L) could reversibly block more than
50% of the intracellular Ca2+ current in rabbits’cardiomyo-
cytes [80]. In isolated rats’cardiomyocytes, tetrandrine (100
micromol/L) reduced Ca2+ influx in the sarcolemma and
inhibited Ca2+ uptake into the sarcoplasmic reticulum by
inhibiting ATP2A2 [81]. Tet (50 mg/kg/d, intragastrically
administrated for nine weeks) [82]) significantly inhibited
calcium overload by reducing the density and the total num-
ber of dihydropyridine binding sites in the myocardium and
vessels. Tet also improved left ventricular compliance and
vascular endothelial function.
(3) Sodium Channel. Tet (10-30 μmol/L) inhibited sodium
current in single bullfrog cardiac cells [83]. Tet (40-
120 μmol/L) inhibited sodium current in cardiomyocytes
from 12 patients with atrial fibrillation without affecting
the density and properties of sodium channels [84]. The
4 Cardiovascular Therapeutics
voltage-gated cardiac sodium channel contributed to action
potential conduction [22]). Dysfunction of sodium channels
increased susceptibility to atrial fibrillation.
3.2.3. Matrine. Extracted from the dried root of Sophora
flavescent Ait. and Euchresta japonica, matrine has antiar-
rhythmia [85], antibacterial [56], antipulmonary [86],
hepatic fibrosis [87], and antitumor [88] effects. As a broad-
spectrum antiarrhythmia drug, matrine can prolong action
potential duration and effective refractory period, slow down
heart rate, improve myocardial contractility, and inhibit
ectopic rhythm and atrioventricular reentry agitation by inhi-
biting potassium, sodium, and calcium channels [89].
(1) Potassium Channel. The HERG gene (human ether-a-go-
go-related gene), also known as KCNH2, was responsible for
encoding Kv11.1 protein [90]. This potassium ion channel
activates the cardiac delayed rapid-rectifying potassium
current (IKr), which was responsible for action potential
platform and 3-phase repolarization [91, 92]. IKr, IKs, and
IK1 were repolarization reserve currents [93].
Matrine had a bidirectional regulation effect on the
HERG potassium channel [94]. In vitro, a low concentration
of marine (1 μmol/L) promoted the expression of HERG in
rats’cardiomyocytes. In contrast, a high concentration of
matrine (100 μmol/L) inhibited the expression of HERG,
prolonged the action potential duration and effective refrac-
tory period (ERP) of ventricular myocytes, gradually slowed
down the frequency of spontaneous discharge, and reduced
the incidence of ectopic rhythm [95].
M3 receptor-mediated K+ current (IKM3) has been
found to be a new target for the treatment of atrial fibrillation
in recent years. Pretreatment of matrine (15, 30, and 45 mg/
kg intravenously administrated once a day for 15 days) sig-
nificantly reduced AF incidence rate and duration time in a
dose-dependent manner. Matrine inhibited atrial repolariza-
tion by inhibiting IKM3 current, prolonged the effective
refractory period, and made the effective refractory period
in different parts of the myocardium tend to be the same,
thus blocking the atrioventricular reentry excitation [96].
Expression of the M3 receptor was decreased, and Cav1.2
expression was upregulated on the atrial membrane [94]
Potassium inward rectifier channel Kir2 (encoded
byKCNJ2) was responsible for terminal cardiac repolariza-
tion and resting membrane stability [97]. “Loss-of-
function”or “Gain-of-function”mutations of KCNJ2 gave
rise to atrial fibrillation. Kv 2.1 (encoded by KCNB1) could
be downregulated in myocardial infarction patients and lead
to electrical instability of the post-MI heart [98]. Matrine
(50, 100, and 200 mg/kg, intragastrically administrated for
seven days) [99] upregulated the expression of KCNB1
(encoding Kv 2.1) and KCNJ2 (encoding Kir2.1) in myocar-
dial tissues of rats with myocardial infarction and prevented
the occurrence of arrhythmia after myocardial infarction.
(2) Calcium Channel. Matrine (15, 30, and 45 mg/kg intrave-
nously administrated once a day for 15 days) upregulated
Cav1.2 expression on atrial membrane. It promoted the
increase of L-type calcium current and the recovery of
calcium-induced calcium release (CICR), which ultimately
improved myocardial contractility and cardiac function
and prevented heart failure in rats with AF [100]. Moreover,
matrine (100 mg/kg/d, intragastrically administrated for four
weeks) improved atrial fibrosis and reduced the susceptibil-
ity of AF in rats with myocardial infarction by inhibiting
the proliferation, migration, and differentiation of cardiac
fibroblasts [101].
(3) Sodium Channel. Voltage and concentration-depen-
dently, matrine inhibited sodium channels. In vitro, matrine
(10, 50, and 100 μmol/L) inhibited sodium current in rat’s
ventricular myocytes and reduced the action potential
amplitude (APA) [102, 103].
3.2.4. Dauricine (Dau). Dauricine is a kind of bibienyl
isoquinoline alkaloid extracted from the roots of Menisper-
mum dauricum DC., with antiarrhythmia [104], anti-
inflammatory [105], antitumor [106], and anticoagulation
[107] effects. In terms of antiarrhythmia, dauricine reduced
Ca2+ and Na+ influx and K+ outflow. Dauricin e (200 μg/
mL) [108] prolonged the atrial effective refractory period
and action potential duration and significantly inhibited
Na+, K+, and Ca2+ current in myocardial tissues.
(1) Potassium Channel. In guinea pig ventricular myocytes,
dauricine (1, 3, 10, 30, and100 μmol/L) inhibited the rapidly
and slowly activating component of the delayed rectifier
potassium current and the inward rectifier potassium cur-
rent [85]. Dau inhibited both active and inactive states of
HERG channels [109]. Unlike quinidine and dofetilide,
dauricine did not affect the deactivation process of Ikr and
Iks and was not likely to cause torsade de pointes ventricular
tachycardia [110].
(2) Calcium Channel. Dauricine was an L-type calcium
channel blocker that reduced intracellular Ca2+ concentra-
tion by inhibiting Ca2+-ATPase activity and sarcoplasmic
reticulum calcium uptake. In rabbit papillary cardiomyo-
cytes, dauricine (30 μmol/L) inhibited early depolarization
by inhibiting L-type calcium channels [111, 112]. Dauri-
cine’s inhibiting effects of Ca2+ channels were also associ-
ated with activated Na+-K+-ATPase and Ca2+-Mg2
+-ATPase [113].
(3) Connexin. Decreased expression or function of connexin
40 protein promoted the aggravation of paroxysmal atrial
fibrillation into persistent atrial fibrillation [114]. Dauricine
(intravenous injecting 5 mg/kg, 30 min before rapid atrial
pacing) inhibited the degradation of Cx40 and the damage
of atrial myocytes caused by rapid atrial pacing [115].
3.2.5. Guanfu Base A (GFA). Extracted from the roots of
Aconitum coreanum (Levl.) Rapaics of Ranunculaceae,
GFA has antiarrhythmia [116], anti-inflammatory [117],
and antioxidation [118] effects. At present, it has been
approved in the treatment of supraventricular tachycardia
in China. In terms of antiarrhythmia, GFA is a kind of
5Cardiovascular Therapeutics
multichannel blocker, which mainly blocks sodium chan-
nels. GFA can not only restrain action potential amplitude
(APA) and Vmax and prolong action potential duration
and effective refractory period but also change unidirectional
conduction block into bidirectional conduction block and
reduce the occurrence of premature contraction and atrio-
ventricular reentry [119, 120].
(1) Potassium Channel. GFA mainly inhibited slow-activated
delayed rectifier potassium current (Iks), with little influence
on rapid-activated delayed rectifier potassium current (Ikr)
[121]. Therefore, it was less likely to cause other arrhythmias
[122]. In a frequency-dependent manner, GFA (100, 400,
1000, and 2500 μmol/L) inhibited potassium currents by
binding to the S6 region of the HERG channel without
affecting the synthesis of HERG proteins. It inhibited the
expression of HERG proteins at high concentrations [123].
GFA did not affect the inward potassium current channel
and transient outward potassium current channel, so it
would not lead to early repolarization [124].
(2) Calcium Channel. By reducing Ca2+ influx, GFA reduced
the depolarization rate and average repolarization rate of
cardiomyocytes [125]. GFA mainly acted on the inactive
state of the L-type calcium channel, which prolonged recov-
ery time from the inactivation state. GFA also had a certain
effect on the calcium channel in the inactive state [126]. GFA
(25, 125, 250, and 1000 μmol/L) blocked L-type calcium
channel in rats’ventricular myocytes in a concentration-
dependent manner.
(3) Sodium Channel. By inhibiting the sodium channel, GFA
reduced the heterotopic automaticity of atrial and ventricu-
lar cells [117]. GFA not only reduced the occurrence of reen-
try by slowing down the atrioventricular bypass conduction
but also prolonged the action potential duration and effec-
tive refractory period. In vitro, GFA (500 μmol/L) reduced
the depolarization rate and average repolarization rate of
rabbits’sinus node cells [127].
In addition, GFA could inhibit the late sodium channel,
which was considered as a potential drug target of AF [128].
Late sodium current increased intracellular sodium and
calcium loading [98] and enhanced susceptibility to atrial
fibrillation [104], heart failure [129], and hypoxia [130].
Mutation in SCN5A (sodium voltage-gated channel alpha
subunit 5) gene increased late sodium current and gave rise
to malignant arrhythmia with pleomorphic ventricular
tachycardia and torsade de pointes (TdP). By inhibiting the
expression of the SCN5A, GFA (100 μmol/L) shortened the
recovery time of action potential and restrained the triggered
activity caused by early after depolarization and delayed
after depolarization [131].
(4) Connexin. GFA (6 and 12 mg/kg, intragastrically admin-
istrated for four days) shortened the duration of atrial fibril-
lation in calcium chlorine-acetylcholine model rats,
prolonged the effective refractory period, reduced the
expression of NADPH oxidase-related subunits, and pro-
moted the communication junction protein expression (con-
nexin 40). In this way, it inhibited atrial electrical
remodeling caused by atrial fibrillation and increased the
success rate of conversion to sinus rhythm.
In a clinical trial, 41 patients with atrioventricular reen-
trant tachycardia in the treatment group were intravenously
injected GFA (200 mg, if the first dose is ineffective, the sec-
ond dose may be given after 15 minutes), and 41 patients in
the control group intravenously were injected propafenone
hydrochloride (70 mg, intravenously injected, if the first dose
is ineffective, the second dose may be given after 30 minutes)
[132]. The results showed that the effective rate of GFA
hydrochloride in the treatment of atrioventricular reentrant
tachycardia was higher than that of propafenone hydrochlo-
ride (87.8% vs. 68.3%, P<0:05). The recovery time of collat-
eral retrograde transmission in the GFA group was longer
than that in the propafenone group (36:6±9:7vs. 19:2±
7:3min, P<0:05).
3.2.6. Neferine. Extracted from the seeds of lotus of Nym-
phaeaceae, neferine has antiarrhythmia, antihypertensive
[133], antitumor [134], antiapoptotic, antioxidative, [135],
anti-ischemic [136], antiallergic, and anti-inflammatory
effects [137]. In terms of arrhythmia, by reducing the Na+,
Ca2+ ion influx, and K+ outflow, nephrine can effectively
prolong the Q-T interphase and slow down heart rate [138].
(1) Potassium Channel. By blocking the HERG potassium
channel, neferine (10 and 30 μmol/L) inhibited potassium
outflow during repolarization, prolonged the action poten-
tial duration, and effective refractory period [116, 140]. In
vitro, neferine (10 and 100 μmol/L) reduced the peak value
of potassium current in rabbit ventricular myocytes by
56.96% and 73.61%, respectively [65].
(2) Calcium Channel. Concentration dependently, neferine
blocked the L-type calcium channel and had a synergistic
effect with verapamil. In vitro, neferine reduced the peak cal-
cium current of rabbit ventricular myocytes by 17.46% and
51.06% at concentrations of 10 and 100 μmol/L (Zuo et al.,
2021 [140]). Neferine inhibited intracellular calcium influx
induced by adenosine diphosphate (ADP) by inhibiting
Ca2+ influx and internal Ca2+ discharge [141].
(3) Sodium Channel. Neferine (30 μmol/L) inhibited activa-
tion of Nav1.5 currents in a frequency-dependent manner
[38] in HEK293 cells. Neferine (40 μmol/L) reduced the
action potential amplitude (APA), maximum rising rate
(Vmax), and maximum rate of depolarization in rats’
cardiomyocytes [142]. Neferine significantly inhibited the
function of the sinoatrial node and the conduction of electri-
cal current between atrial and ventricular cells [110].
3.3. Quinones
3.3.1. Tanshinone IIA. Extracted from the root of Salvia
miltiorrhiza Salvia miltiorrhiza Bunge., tanshinone?A has
anti-arrhythmic [143], anticoagulant, anti-ischemic [144],
6 Cardiovascular Therapeutics
anti-neoplastic [145], and anti-inflammatory [146] effects.
By inhibiting potassium and calcium ion channels, tanshi-
none IIA can prolong the effective refractory period and
action potential duration, inhibit cardiac repolarization,
and increase the threshold of ventricular fibrillation [147].
(1) Potassium Channel. The KCNJ2 gene that encoded
Kir2.1 and Kir2.2 proteins of inward rectifier potassium
channels [121] was associated with the pathogenesis of atrial
fibrillation [148]. KCNQ1 gene-encoded Kv7.1 protein of
Iks, while KCNE1 encoded Mink protein of Iks channel. In
myocytes of AF rats, tanshinone IIA upregulated the expres-
sion of KCNJ2, downregulated the expression of KCNQ1
and KCNE1, and inhibited myocardial cell potassium out-
flows and repolarization of cardiomyocytes [129, 149]. In
vivo, tanshinone IIA (2 mg/kg) significantly prolonged the
rabbit ventricular relative refractory period, effective refrac-
tory period [150]. Because of little impact on the Ikr channel,
tanshinone IIA was less arrhythmogenic than sotalol.
(2) Calcium Channel. Tanshinone IIA (32 mg/kg, intragastri-
cally administrated for 14 days) significantly upregulated the
expression of the CACNA1C gene in atrial tissue of AF rats
and improved atrial electrical remodeling and calcium over-
load [41]. Tanshinone IIA reduced the expression of
microRNA-1 through the p38 mitogen-activated protein
kinase pathway [129, 151]. In addition, tanshinone IIA
inhibited collagen secretion induced by AngII and the syn-
thesis rate of atrial fibroblasts by means of inhibiting the
TSP-1/TGF-β1 pathway [152]. The differentiation of atrial
fibroblasts into myofibroblasts plays an important role in
atrial fibrosis. As a water-soluble derivative of tanshinone
IIA, sodium tanshinone IIA sulfonate prevented atrial fibro-
sis by inhibiting oxidative stress and TGF-βactivation in the
AngII-1 signaling pathway [153].
In China, tanshinone IIA has been widely used in the
clinical treatment of myocardial infarction complicated by
atrial fibrillation. Wang and Xie [154] conducted a meta-
analysis that included ten clinical studies (involving 1088
patients). The results showed that success rate of conversion
into sinus rhythm (OR = 3:10, 95%CI = 2:16-4.44, P<0:05),
the recurrence rate of atrial fibrillation (OR = 0:32,
95%CI = 0:22-0.48, P<0:05), the incidence rate of heart fail-
ure (OR = 0:23, 95%CI-0.15-0.35, P<0:05), mortality
(OR = 0:23,95%CI = 0:15-0.35, P<0:05), and adverse reac-
tion rate (OR = 0:24, 95%CI = 0:12-0.49, P<0:05) in the
treatment group (Tanshinone IIA sulfonate combined with
amiodarone) were better than those in the control group
(only amiodarone).
3.4. Polyphenols. Polyphenols are secondary metabolites with
polyatomic phenol structures that are widely found in the
skin, roots, leaves, and fruits of medicinal plants like Polyg-
onum cuspidatum Sieb.et Zucc., Syringa oblata Lindl., and
Paeonia suffruticosa Andr. Among them, resveratrol, puer-
arin, and acacetin have been found with good antiarrhyth-
mic effects.
3.4.1. Resveratrol. Extracted from dry roots of Polygonum
cuspidatum Sieb.et Zucc. of Polygonaceae, resveratrol has
antiarrhythmia [155], antioxidation [156], antitumor [157],
anti-ischemic [158], and antiplatelet [159] effects. In terms
of antiarrhythmic, resveratrol can slow down the heart rate,
prolong the effective refractory period of cardiomyocytes,
and inhibit the occurrence of early and delayed after depo-
larization [158], by regulating potassium, calcium, and
sodium ion channels [160].
(1) Potassium Channel. Resveratrol prolonged action poten-
tial duration and effective refractory period by inhibiting the
expression of the HERG gene, as well as rapid and slow acti-
vation of delayed rectifying potassium ion current [161]. In
vitro, resveratrol (50, 100, and 500 μM) slowed down guinea
pigs’heart rate and inhibited myocardial contractility in a
dosage-dependent manner by regulating ATP-sensitive
potassium channels, transient outward potassium current,
calcium-activated potassium channels, and inward rectifying
potassium channels [162].
Resveratrol enhanced Kv2.1 potassium current in H9C2-
rat cardiomyocytes in a time- and concentration-dependent
manner. The median maximum effective concentration was
14.02 μmol/L [163].
(2) Calcium Channel. Resveratrol (1, 50, and 100 μmol/L)
inhibited the L-type calcium channel, reduced the intracellu-
lar calcium influx, and prolonged the effective refractory
period in guinea pig ventricular myocytes [164, 165]. Resver-
atrol not only inhibited the occurrence of early and delayed
after depolarization (EAD and DAD) but also slowed down
the atrioventricular conduction and inhibited atrioventricu-
lar node reentry excitement. In vitro, resveratrol protected
guinea pigs’ventricular myocytes from oxidative stress-
induced arrhythmias and calcium overload, by means of
inhibiting L-type calcium channel, reducing the production
of oxygen free radicals in cardiomyocytes, and preventing
the activation of calmodulin-activated protein kinase II
(CaMK II) [155].
(3) Sodium Channel. Concentration-dependently, resveratrol
(10, 30, and 100 μmol/L) inhibited the late sodium current
and the reverse type sodium-calcium exchangers of ventric-
ular myocyte of guinea pig [166]. At the concentration of
100 μmol/L, resveratrol could inhibit 52:7±10:2%of
sodium current [167]. Resveratrol (10, 20, 40, and 80 μM)
inhibited ischemic arrhythmias by inhibiting the H2O2-
induced late sodium current and the reverse sodium-
calcium exchange current (INCX) [168].
(4) Connexin 43. Resveratrol could regulate the expression of
the GJA gene and myocardial connexin 43 (Cx43), which
play a therapeutic role in idiopathic atrial fibrillation [169].
Resveratrol (1 mL/kg, intravenous ly administered) upregu-
lated the expression and activity of Cx43 in male SD rats
by activating the PI3K/Akt signaling pathway, thus prevent-
ing myocardial ischemia-reperfusion arrhythmia [170]. Res-
veratrol (2.5 mg/kg/d, intragastrically administrated for
7Cardiovascular Therapeutics
seven days) inhibited atrial remodeling and reduced AF by
increasing activity of deacetylase 1 (SIRT1) [171].
(5) HCN Channel. Resveratrol inhibited hyperpolarization-
activated cyclic nucleotide-gated cation channel 4 (HCN4)
(IC50 value = 83:75 μmol/L) [172]. Resveratrol inhibited
Kv1.5 current and IKAch [173] frequency-dependently, with
IC₅₀ of 0.36 and 1.9 μmol/L [173].
3.4.2. Puerarin. Extracted from the dry root of Pueraria
puerariae, puerarin is a kind of flavonoid compound with
antiarrhythmia [175], anti-ischemic [176], antihypertensive
[177], hypolipidemic [178], hypoglycemic [179], coronary
vasodilation [180], and anti-inflammatory [181] effects. In
China, puerarin (Yufeng Ningxin Dropping Pill) has been
approved for the clinical treatment of hypertension, coro-
nary heart disease, angina pectoris, and neuropathic head-
ache. In terms of antiarrhythmia, puerarin can slow down
the heart rate, inhibit the cardiomyocyte automaticity, pro-
long the effective refractory period, and action potential
duration [182, 183].
(1) Potassium Channel. Puerarin (0.01, 0.1, and 1 mmol/L)
prolonged the duration of the action potential by inhibiting
IKs in rat ventricular myocytes [184]. Zhang et al. [185]
found that puerarin was a novel antagonist towards inward
rectifier potassium channel (IK1). Puerarin (1.2 mmol/L)
significantly inhibited the IK1 current in rat ventricular cells.
In the transfected HEK293 cells, puerarin significantly
inhibited inward rectifying K+ channels in a dose-
dependent manner and had a more significant effect on
Kir2.1 and Kir2.3. Puerarin significantly inhibited Kv7.1
and IKs in micromolar concentrations [186]. Moreover, by
regulating the calcium-activated potassium channel and
activating protein kinase C, puera rin (0.24 mmol/L) pro-
tected rats’ventricular myocytes against ischemia and reper-
fusion injury [187].
(2) Calcium Channel. Puerarin (2.4 mmol/L) inhibited the L-
type calcium channel in rats’ventricular myocytes in a time-
dependent manner [188]. Puerarin (100 mg/kg, iv) [189] sig-
nificantly inhibited the arrhythmias (including ventricular
premature contraction, ventricular tachycardia, and ventric-
ular fibrillation) induced by chloroform (2 mL inhaled by
mice) or aconitine (40 mg/L intravenous injected to SD rats).
Puerarin ([190], 100 mg/kg intraper itoneally injected for ten
days) inhibited isoproterenol-induced cardiac hypertrophy
and reduced intracellular calcium concentration.
(3) Sodium Channel. M. Puerarin inhibited sodium channels
in a dose-dependent manner with IC ð50Þ= 349 μmol/L
[191]. Wang conducted a prospective cohort study involving
87 patients with persistent atrial fibrillation. Forty-three
patients in the control group received amiodarone (0.2 g
tid for one week, 0.2 g, bid for one week, and 0.2 g QD for
the last week); 44 patients in the combination group received
puerarin (40 mL + 250 mL 0.9% NaCl, ivgtt, for 21 days)
based on the control group. The results showed that the
average time of restoring sinus rhythm in the combination
group was significantly shorter than that of the control
group (7.5 d vs. 10.2 d, P<0:01), and the success rate of
restoring sinus rhythm was significantly higher than that of
the control group (77.3% vs. 60.5%, P<0:01)[192].
3.4.3. Acacetin. Acacetin is a kind of flavonoid extracted from
Saussurea involucrata (Kar. et Kir.) Sch.-Bip., with antiar-
rhythmic [193], antitumor [194, 195], anti-inflammatory
[196], antifibrosis [197], and antioxidation [198]. In terms
of antiarrhythmia, acacetin is a kind of potassium channel
inhibitor without effect on the Na(+) and L-type Ca (2+)
channels. Acacetin (3, 6, and 12 mg/kg, intravenously admin-
istrated for seven days) terminated experimental AF of beagle
dogs without prolonging QTc interval [199].
(1) Potassium Channel. Acacetin (5–10 μM) reduced Ito
density, action potential notch, and J wave area in electrocar-
diograph [200]. Encoded by KCNA5, ultrarapid delayed
rectifier potassium channel (Kv1.5) was only expressed in
human atrial myocytes. Inhibiting the Kv1.5 current could
prolong atrial action potential duration and increase the
refractory period of the fibrillating atrium [14]. Acacetin
(IC50 = 3:2and 9.2 μmol/L, respectively) inhibited ultra-
rapid delayed rectifier potassium current I (Kur) and the
transient outward K+ current and IKACh in human atrial
myocytes [201], with little potency in inhibiting IKr and
IKs. Other flavonoids, including hesperetin [14], myricetin
[202], and quercetin [203], could also inhibit I (Kur).
Blocking the K+ channel could enhance the AF-
selectivity of INa blockade. As a K+-current blocker, acacetin
combined with INa blocker showed synergistic antiarrhyth-
mic benefits without significant alterations of ventricular
repolarization and QT intervals.
Acacetin showed a weak inhibition in the hERG and
KCNQ1/KCNE1 channels [193] in rabbit hearts. In cardio-
myocytes of anesthetized dogs, acacetin (2.5, 5, and 10 mg/
kg) prevented AF induction [193]. Chen et al. [204] found
that acacetin inhibited small conductance Ca2+-activated K
+ channel (SKCa) currents with IC50 of 12.4 μM for SKCa1,
10.8 μM for SKCa2, and 11.6 μM for SKCa3.
3.5. Organic Acid. Organic acids are organic compounds
with acidity that are widely distributed in leaves, roots,
and fruits of Chinese Materia Medica like Glycyrrhiza
uralensis Fisch.
3.5.1. Glycyrrhizic Acid (GA). Extracted from the dried root
and rhizome of Glycyrrhiza uralensis Fisch., Glycyrrhiza
inflata Bat., or Glycyrrhiza glabra L., glycyrrhizic acid has
antiarrhythmia [205], antioxidant, anti-inflammatory [206],
antiviral [181], and antitumor (Zuo et al., 2021) effects
[207]. In terms of arrhythmia, glycyrrhizic acid can block
sodium and calcium ion channels to inhibit the automaticity
of pacemaker cells, slow down the conduction speed,
prolong the action potential duration, and effective refrac-
tory period [208].
8 Cardiovascular Therapeutics
Table 1: The effects of active components of antiarrhythmic natural products exerted on ion channels.
Origin Names Pharmacological
action Chemical structure Potassium channel Calcium
channels
Sodium
channel Connexin HCN
Panax
notoginseng
(Burk.) F.H.
Chen
Panax
notoginseng
saponins
Antiarrhythmia [42],
antiplatelet [37],
antishock [39],
antioxidation [225],
sedative [40], and
antitumor [41]
R3O
R1O
R2
OH
Small conductance
calcium activates
potassium channel,
potassium calcium-
activated channel
[43]
L-type and
T-type Ca2+
channel [44]
[50]
Coptis
chinensis
Franch.,
Phellodendron
chinense
Schneid,
Mahonia
bealei (Fort.)
Carr
Berberine
Antiarrhythmia [54],
anti-inflammatory
[55], antibacterial
[56], hypoglycemia
[57], vasodilation
[58], antitumor
effects [59].
O
O
O
O
N+
Delayed rectifier
outward potassium
channel, ATP-
sensitive K+ (KATP)
channels, inward
rectifier potassium
channel ([64, 65])
L-type and
T-type
calcium
channel,
ryanodine
receptor,
sarcoplasmic/
endoplasmic
reticulum
calcium
ATPase
([66]; [67])
[68]
hHCN4 [70]
Stephania
tetrandra S.
Moore.
Tetrandrine
Antiarrhythmia [73],
antihypertensive
[74], anti-
inflammatory [75],
and antitumor [74]
OO
O
O
O
NN
O
Delayed rectifier
potassium channel,
calcium-activated
potassium channel
[18]
L-type and
T-type
calcium
channels,
sarcoplasmic/
endoplasmic
reticulum
calcium
ATPase [80]
[82]
Voltage-
gated
cardiac
sodium
channel
[22] [84]
Sophora
alopecuroides
L., Sophora
flavescens Alt.
Matrine
Antiarrhythmia [85],
antibacterial [226],
antipulmonary [86],
and hepatic fibrosis
[87], and antitumor
[88]
N
N
ODelayed rectifier
potassium channel,
M3 receptor-
mediated K+
channel [96] [94],
potassium inward
rectifier channel [98]
[99]
L-type
calcium
channels
[100] [101]
Voltage-
gated
cardiac
sodium
channel
([103];
[102])
9Cardiovascular Therapeutics
Table 1: Continued.
Origin Names Pharmacological
action Chemical structure Potassium channel Calcium
channels
Sodium
channel Connexin HCN
Menispermum
dauricum DC.Dauricine
Antiarrhythmia
[104], anti-
inflammatory [105],
antitumor [106],
anticoagulation
[107]
O
OOH
O
O
O
N
N
Delayed rectifier
potassium channel,
inward rectifier
potassium channel
[94] [95] [96]
L-type
calcium
channels
[101] [100]
Cx40
([27]; [28])
[29] [30]
[31] [115]
Aconitum
coreanum
(Levl.) Rapaics
Guanfu base
A
Antiarrhythmia
[116], anti-
inflammatory [117],
and antioxidation
[118]
O
O
N
O
O
HO
OH
Delayed rectifier
potassium channel
[121] [122] [124]
L-type
calcium [125]
[126]
Voltage-
gated
cardiac
sodium
channel
[117] [127]
Cx40 [132]
Lotus of
Nymphaeaceae Neferine
Antiarrhythmia,
antihypertensive
[133], antitumor
[134], antiapoptotic
and antioxidative
[135], anti-ischemic
[136], antiallergic,
and anti-
inflammatory
effects[137]
O
O
N
O
N
O
HO
O
HERG potassium
channel ([116];
[140])
L-type
calcium
channel (Zuo
et al., 2021)
[141]
Voltage-
gated
cardiac
sodium
channel
[38] [142]
[110]
10 Cardiovascular Therapeutics
Table 1: Continued.
Origin Names Pharmacological
action Chemical structure Potassium channel Calcium
channels
Sodium
channel Connexin HCN
Salvia
miltiorrhiza
Bge.
Tanshinone
IIA
Antiarrhythmic
[143],
anticoagulation,
anti-ischemia [144],
antitumor [145], and
anti-inflammation
[146]
O
OO
Delayed rectifier
potassium channel
([129]; [149]),
inward rectifier
potassium channel
[227] [148]
type calcium
channel [41]
Voltage-
gated
cardiac
sodium
channel
[152]
Vitis vinifera
L., Punica
granatum L.,
Vaccinium
spp.,
Vaccinium
macrocarpon
Resveratrol
Antiarrhythmia
[155], antioxidation
[156], antitumor
[157],
antimyocardial
ischemia [158], and
antiplatelet [159]
HO
HO
OH Delayed rectifier
potassium channel
[161], ATP-sensitive
potassium channels,
transient outward
potassium channel,
calcium-activated
potassium channel,
and inward
rectifying potassium
channel [163]
L-type
calcium
([164]; [165])
[155]
Late
sodium
channel
and reverse
type
sodium-
calcium
exchangers
[166]
Cx43 [169]
[170]
Hyperpolarization-
activated cyclic
nucleotide-gated
cation channel
[172] [173]
Pueraria
lobata (Willd)
Ohwi,
Pueraria
thunbergiana
Benth.
Puerarin
Antiarrhythmia
(Wei et al., 2015
[175]), anti-ischemic
[176],
antihypertensive
[177], lowering
blood lipid [178],
lowering blood sugar
[179], expanding
coronary arteries
[228], and anti-
inflammatory [181]
HO
HO
HO
O
O
O
OH
OH
OH
Delayed rectifier
potassium channel,
inward rectifier
potassium channel
[185] [186], calcium-
activated potassium
channel [187]
L-type
calcium [188]
Voltage-
gated
cardiac
sodium
channel
[191]
Saussurea
involucrata
(Kar. et Kir.)
Sch.-Bip.
Acacetin
Antiarrhythmic
[193], antitumor,
anti-inflammatory,
antifibrosis,
antioxidation [198]
HO
HO
O
O
O
Ultrarapid delayed
rectifier potassium
channel and the
transient outward
potassium channel
[193], hERG, small
conductance Ca2
+-activated K+
channels [204],
transient outward
potassium channel
[200]
11Cardiovascular Therapeutics
Table 1: Continued.
Origin Names Pharmacological
action Chemical structure Potassium channel Calcium
channels
Sodium
channel Connexin HCN
Glycyrrhiza
uralensis
Fisch.,
Glycyrrhiza
inflata Bat., or
Glycyrrhiza
glabra L.
Glycyrrhizic
acid
Antiarrhythmia
[205], antioxidant,
anti-inflammatory
[206], antiviral
[229], antitumor
(Zuo et al., 2021)
O
OH
HO
HO
HO
HO
HO
OH
OH
O
O
O
O
O
O
O
L-type
calcium
channel
[205]
Voltage-
gated
cardiac
sodium
channel
[210] [211]
Artemisia
carvifolia
Buch.-Ham. ex
Roxb. Hort.
Beng.
Artemisinin
Antimalarial [212],
antitumor [213],
anti-inflammatory
[214], antiviral
[215], antibacterial
[216], and
antifibrotic [217]
O
O
O
O
O
Delayed rectifier
potassium channel,
inward rectifier
potassium channel,
transient outward
channel [218] [219]
L-type
calcium
channel
[220]
Cx43[221]
[222]
Hyperpolarization-
activated cyclic
nucleotide-gated
cation channel
[223]
12 Cardiovascular Therapeutics
(1) Calcium Channel. Glycyrrhizic acid can act on the CAC-
NA1C gene to inhibit the L-type Ca2+ channel current in
cardiomyocytes. Therefore, it can hinder sinus node pacing
early and delayed depolarizing triggering activities [205].
Adrenergic receptors of the heart stimulated by epineph-
rine and norepinephrine can promote the release of cAMP,
phosphoinositide, and the second messenger signaling cas-
cade [209]. In vitro, ten μM GA can inhibit the cAMP levels
of CHO cells transfected with β2-AR or β3-AR, suggesting
its’selective antagonistic capability against β2-AR and β3-
AR [205].
(2) Sodium Channel. Glycyrrhizic acid can inhibit sodium
ion channels in cardiomyocytes in a concentration-
dependent manner [210]. Glycyrrhizic acid can inhibit Na
+influx of cardiac myocytes during depolarization, reduce
action potential amplitude and maximum rise rate of the
action potential, slow down conduction velocity, and reduce
Na + influx in phase 4 of action potential duration in ectopic
pacemaker cells, thus decreasing the excitability of ectopic
pacers [211].
3.6. Terpenoids. Widely existing in natural products, terpe-
noids are compounds and derivatives derived from methyl
glutaric acid and whose molecular skeleton takes isoprene
unit as the basic structural unit.
3.6.1. Artemisinin. Extracted from Artemisia annua L., arte-
misinin has antimalarial [212], antitumor [213], anti-
inflammatory [214], antiviral [215], antibacterial [216], and
antifibrotic [217] effects. Artemisinin (5, 10, and 20 mg/kg,
intraperitoneally injected for once) inhibited arrhythmia
induced by barium chloride by regulating sympathetic tone.
Artemisinin was superior to amiodarone in prolonging QT
interval (4.0 mg/kg, intraperitoneally injected), and there
were fewer adverse reactions [180].
(1) Potassium Channel. Artemisinin (5 and 50 μmol/L) inhib-
ited the inward rectifier potassium channel (IK1) of African
frog [218] and inhibited IK1, It0, and delayed activation recti-
fier potassium current of dogs’cardiomyocytes [219].
(2) Calcium Channel. Artemisinin (4, 2, and 1 mg intragas-
trically administrated for 28 days) upregulated Cav1.2
calcium channel expression level, downregulated the expres-
sion level of calmodulin and calmodulin-dependent protein
kinase II (CaMKII), and inhibited the level of phosphory-
lated ryanodine receptor 2 [220].
(3) Connexin 43. Cx43 remodeling was associated with
action potential duration dispersion and reducing conduc-
tion [221]. Artemisinin increased Cx43 expression by
inhibiting TNF-αand attenuating sympathetic tone [222].
(4) Hyperpolarization-Activated Cyclic Nucleotide-Gated
Cation Channel (HCN). HCN mainly had the following
characteristics: activated by hyperpolarization, sodium-
potassium ion mixed channel, double regulated by voltage,
and cyclic nucleotide [223]. HCN channel was the primary
determinant of phase 4 automatic depolarization of auto-
rhythmic cells and improved the autonomy of autorhythmic
Matrine PNS PNS
Berberine
Berberine
Tetrandrine
PNS
ATP2A2
HCN
RyR2
Berberine
Berberine
Tetrandrine
PNS
Berberine
Tetrandrine
Matrine
Guanfu
base A
Berberine
Tetrandrine
Guanfu base
A
Guanfu base A
Dauricine
Dauricine
TanshinoneIIA
TanshinoneIIA
TanshinoneIIA
Resveratrol
Resveratrol
Resveratrol
Resveratrol
Resveratrol
Puerarin
Puerarin
Puerarin
Artemisinin
Artemisinin
Berberine
Tetrandrine
Tetrandrine
Matrine
Matrine
Dauricine
Artemisinin
Artemisinin
Artemisinin
Neferine
Neferine
Neferine
Glycyrrhizic
acid
Glycyrrhizic
acid
IKs
IKr
IK
IK Ach
INa
IK ATP IKM3 ISK
ICa, T
ICa, L
Figure 2: Natural products’effects on potassium, sodium, and calcium channels.
13Cardiovascular Therapeutics
cells. Artemisinin (75 mg/kg, intragastrically administrated,
three times a day for four weeks) inhibited the pacing cur-
rent of sinoatrial node in rabbits with heart failure, reduced
the expression of HCN channel, and thus reduced the heart
rate [224].
4. Summary
Based on the classification of chemistry structure, the effects
of saponins, alkaloids, polyphenols’effects on potassium,
sodium, and calcium channels of cardiomyocytes are
summarized in this article (see Table 1 and Figure 2). In
addition, the natural products’regulatory effects exerted on
the expression level and function of genes responsible for encod-
ing ion channel protein are also summarized (see Table 2).
However, most of the studies above are limited to animal
or cell experiments, which only preliminarily demonstrate
their therapeutic effect on atrial fibrillation. In the future,
studies on pharmacokinetics/pharmacodynamics and mech-
anisms as well as clinical trials of natural products with anti-
arrhythmic activity should be performed. We will continue
to track relevant reports to promote the development of
new AADs.
5. Conclusion
Potassium, calcium, sodium channel, connexins, and HCN
channel are involved in the pathogenesis of atrial fibrillation.
In terms of potassium channels, predominantly expressed in
atria, ultrarapidly delayed rectifier potassium channel (Kur)
and small conductance calcium-activated potassium channel
(SKCa) can be considered to be atrial-selective targets for
developing anti-AF drugs. Kur can enhance spiral-wave
reentry. SKCa can prolong atrial repolarization in the
pulmonary vein and inhibit the maintenance of atrial fibril-
lation. Ectopic rhythm caused by EAD, DAD, and calcium
overload can be inhibited by blocking L-type calcium chan-
nels or late sodium channels. Increasing the expression or
function of connexins can inhibit reentry and heterogeneous
conduction. HCN channel is a promising channel in heart
rate control therapy by inhibiting diastolic depolarization.
Tanshinone IIA and Guanfu A have been approved in
the clinical treatment of arrhythmia in China. The advantage
of tanshinone IIA is inhibiting structural and electrical
remodeling. The benefit of Guanfu A is inhibiting triggered
activity and atrioventricular reentry. Although PNS, berber-
ine, and puerarin have been found effective in clinical trials
of paroxysmal atrial fibrillation, their efficacy still needs to
be confirmed by well-designed, randomized, double-blind
controlled trials.
Abbreviations
AAD: Antiarrhythmic drugs
CNKI: Chinese National Knowledge Infrastructure
AF: Atrial fibrillation
NICE: National Institute for Health and Care
Excellence
AHA: American Heart Association
ACC: American College of Cardiology
HRS: Heart Rhythm Society
EACTS: European Association of Cardio-Thoracic
Surgery
KCNA5: Potassium voltage-gated channel subfamily A
member 5
KCNN3: Potassium calcium-activated channel subfam-
ily N member 3
miR-499: MicroRNA-499
Table 2: Natural product active components’regulatory effects on the genes responsible for ion channel proteins.
Gene Encoding protein Upregulate Downregulate
SCN5A Nav1.5 GFA
KCNB1 Kv2.1 Matrine
KCND3 Kv4.3 PNS
KCNE1 Mink Tanshinone IIA
KCNH2 Kv11.1 Matrine Berberine, matrine,
resveratrol
KCNJ2 Kir2.1 Matrine, tanshinone IIA
KCNQ1 Kv7.1 Tanshinone IIA
KCNN3 KCa2.3 PNS
hHCN4 Hyperpolarization-activated cyclic nucleotide gated potassium
channel Berberine
SK3 Small conductance calcium activates potassium channel 3 PNS
CACNA1C Cav1. 2 Matrine, Tanshinone
IIA PNS
CACNB1 Calcium voltage-gated channel auxiliary subunit beta PNS
RyR2 Ryanodine receptor Berberine
GJA1 Gap junction protein 43 (Cx43) Resveratrol
GJA5 Gap junction protein 40 (Cx40) GFA, puerarin
ATP2A2 ATPase sarcoplasmic/endoplasmic reticulum Ca2+ transporter PNS, berberine
14 Cardiovascular Therapeutics
miR-29: MicroRNA-29
IKr: Delayed rapid-rectifying potassium current
IKM
3
: M3 receptor-mediated K
+
current
CICR: Calcium-induced calcium release
SCN5A: Sodium voltage-gated channel alpha subunit 5
KCNB1: Potassium voltage-gated channel subfamily B
member 1
PI3K: Phosphatidylinositol 3 kinase
KCNE1: Potassium voltage-gated channel subfamily E
member 1
CACNB1: Calcium voltage-gated channel auxiliary sub-
unit beta 1
GJA1: Gap junction protein alpha 1
GFA: Guanfu base A
Iks: Slow activated delayed rectified potassium
current
KCNQ1: Potassium voltage-gated channel subfamily Q
member 1
TGF-β1: Transforming growth factor beta 1
INCX: Reverse sodium-calcium exchange current
CaM: Calmodulin
IK1: Inward rectifier potassium channel
RyR2: Ryanodine receptor2
SERCA2: Sarcoplasmic reticular Ca
2+
-ATPase
KCNE1: Potassium calcium-activated channel subfam-
ily E member 1
KCNE3: Potassium calcium-activated channel subfam-
ily E member 3
HCN2: Hyperpolarization-activated cation channel 2
KChIP2: Potassium channel interacting protein 2
PNS: Panax notoginseng saponins
APD: Action potential duration
ERP: Effective refractory period
SK3: Small conductance calcium activates potas-
sium channel 3
ATP2A2: ATPase sarcoplasmic/endoplasmic reticulum
Ca
2+
receptor 2
HERG: Human ether-a-go-go-related gene
hHCN4: Hyperpolarization-activated cyclic nucleotide-
gated 4
APA: Action potential amplitude
KCNH2: Potassium voltage-gated channel subfamily H
member 2
KCND3: Potassium voltage-gated channel subfamily D
member 3
KCNJ2: Potassium voltage-gated channel subfamily J
member 2
KCNQ1: Potassium voltage-gated channel subfamily Q
member 1
CACNA1C: Calcium voltage-gated channel subunit alpha1 C
RyR2: Ryanodine receptor 2
GJA5: Gap junction protein alpha 5
Dau: Dauricine
Ikr: Rapid activated delayed rectified potassium
current
TSP-1: Thrombospondin-1
Ang II: Angiotensin II
CaMK II: Calmodulin-activated protein kinase II
Cx43: Connexin 43
SIRT1: Sirtuin 1.
Data Availability
The data used to support the findings in this study are
included within the article.
Disclosure
No funder support was involved in the manuscript writing,
editing, approval, or decision to publish.
Conflicts of Interest
The authors declare that they have no conflicts of interest.
Authors’Contributions
Jinshan He and Sicong Li contributed equally to this arti-
cle. They are both responsible for the initial outline, draft
writing, revisions, and final approval. Yumeng Ding was
responsible for English translation, presentation, draft
writing, and revisions for intellectual content. Xuebin Li
was the corresponding author and responsible for revi-
sions for intellectual content and final approval. Jinshan
He and Sicong Li contributed equally to this work.
Acknowledgments
The authors would like to thank all the scholars who have
made outstanding contributions to natural products.
References
[1] M. Perry, S. Kemmis Betty, N. Downes, N. Andrews, and
S. Mackenzie, “Atrial fibrillation: diagnosis and
management-summary of NICE guidance,”BMJ, vol. 373,
no. 1150, 2021.
[2] C. T. January, L. S. Wann, H. Calkins et al., “2019 AHA/ACC/
HRS focused update of the 2014 AHA/ACC/HRS guideline
for the management of patients with atrial fibrillation: a
report of the American College of Cardiology/American
Heart Association task force on clinical practice guidelines
and the Heart Rhythm Society in collaboration with the Soci-
ety of Thoracic Surgeons,”Circulation, vol. 140, no. 2,
pp. e125–e151, 2019.
[3] R. Schleberger, A. Rillig, P. Kirchhof, A. Metzner, and
B. Reissmann, “Update vorhofflimmern: die ESC-leitlinien
2020 sowie aktuelle daten zur frühen antiarrhythmischen
therapie,”Herzschrittmachertherapie & Elektrophysiologie,
vol. 32, no. 2, pp. 257–263, 2021.
[4] M. J. Mulder, M. Kemme, and C. P. Allaart, “Radiofrequency
ablation to achieve durable pulmonary vein isolation,”Euro-
pace: European pacing, arrhythmias, and cardiac electrophys-
iology: journal of the working groups on cardiac pacing,
arrhythmias, and cardiac cellular electrophysiology of the
European Society of Cardiology, vol. 279, 2021.
[5] F. Fusi, A. Trezza, M. Tramaglino, G. Sgaragli, S. Saponara,
and O. Spiga, “The beneficial health effects of flavonoids on
the cardiovascular system: focus on K
+
channels,”Pharmaco-
logical Research, vol. 152, article 104625, 2020.
15Cardiovascular Therapeutics
[6] C. Li, X. Li, X. Gao et al., “MicroRNA-328 as a regulator of
cardiac hypertrophy,”International Journal of Cardiology,
vol. 173, no. 2, pp. 268–276, 2014.
[7] N. Gaborit, M. Steenman, G. Lamirault et al., “Human atrial
ion channel and transporter subunit gene-expression remod-
eling associated with valvular heart disease and atrial fibrilla-
tion,”Circulation, vol. 112, no. 4, pp. 471–481, 2005.
[8] S. Oh, K. B. Kim, H. Ahn, H. J. Cho, and Y. S. Choi, “Remod-
eling of ion channel expression in patients with chronic atrial
fibrillation and mitral valvular heart disease,”The Korean
Journal of Internal Medicine, vol. 25, no. 4, pp. 377–385, 2010.
[9] P. Biliczki, R. A. Boon, Z. Girmatsion et al., “Age-related reg-
ulation and region-specific distribution of ion channel sub-
units promoting atrial fibrillation in human left and right
atria,”EP Europace, vol. 21, no. 8, pp. 1261–1269, 2019.
[10] D. Dobrev, A. Friedrich, N. Voigt et al., “The G protein-gated
potassium CurrentIK,AChIs constitutively active in patients
with chronic atrial fibrillation,”Circulation, vol. 112, no. 24,
pp. 3697–3706, 2005.
[11] N. Voigt, A. Trausch, M. Knaut et al., “Left-to-right atrial
inward rectifier potassium current gradients in patients with
paroxysmal versus chronic atrial fibrillation,”Circulation.
Arrhythmia and Electrophysiology, vol. 3, no. 5, pp. 472–
480, 2010.
[12] J. Kneller, R. Zou, E. J. Vigmond, Z. Wang, L. J. Leon, and
S. Nattel, “Cholinergic atrial fibrillation in a computer model
of a two-dimensional sheet of canine atrial cells with realistic
ionic properties,”Circulation Research, vol. 90, no. 9,
pp. E73–E87, 2002.
[13] R. Aldrich, K. G. Chandy, S. Grissmer, G. A. Gutman, A. D.
Wei, and H. Wulff,“Calcium-activated potassium channels,
introduction,”IUPHAR/BPS Guide to Pharmacology,
vol. 22, 2015.
[14] N. Schmitt, M. Grunnet, and S. P. Olesen, “Cardiac potas-
sium channel subtypes: new roles in repolarization and
arrhythmia,”Physiological Reviews, vol. 94, no. 2, pp. 609–
653, 2014.
[15] E. Wettwer and H. Terlau, “Pharmacology of voltage-gated
potassium channel K
v
1.5 –impact on cardiac excitability,”
Current Opinion in Pharmacology, vol. 15, pp. 115–121, 2014.
[16] F. Wiedmann, N. Frey, and C. Schmidt, “Two-pore-domain
potassium (K2P-) channels: cardiac expression patterns and
disease-specific remodelling processes,”Cells, vol. 10,
no. 11, p. 2914, 2021.
[17] R. Gardner, I. G. Crozier, A. L. Binfield et al., “Penetrance and
expressivity of the R858H CACNA1C variant in a five-
generation pedigree segregating an arrhythmogenic chan-
nelopathy,”Molecular Genetics & Genomic Medicine, vol. 7,
no. 1, article e00476, 2019.
[18] P. Nieto-Marín, J. Jiménez-Jáimez, D. Tinaquero et al.,
“Digenic Heterozigosity in _SCN5A_ and _CACNA1C_
Explains the Variable Expressivity of the Long QT Phenotype
in a Spanish Family,”Revista espanola de cardiologia, vol. 72,
no. 4, pp. 324–332, 2019.
[19] M. Y. Baek, H. S. Hwang, J. Y. Park et al., “Association
between CACNA1C gene polymorphisms and ritodrine-
induced adverse events in preterm labor patients,”European
Journal of Clinical Pharmacology, vol. 73, no. 7, pp. 837–842,
2017.
[20] D. Dobrev, N. Voigt, and X. H. Wehrens, “The ryanodine
receptor channel as a molecular motif in atrial fibrillation:
pathophysiological and therapeutic implications,”Cardiovas-
cular Research, vol. 89, no. 4, pp. 734–743, 2011.
[21] D. M. Roden, J. R. Balser, A. L. J. George, and M. E. Anderson,
“Cardiac ion channels,”Annual Review of Physiology, vol. 64,
no. 1, pp. 431–475, 2002.
[22] H. Xiong, Q. Yang, X. Zhang et al., “Significant association of
rare variant p.Gly8Ser in cardiac sodium channel β4-subunit
SCN4B with atrial fibrillation,”Annals of human genetics,
vol. 83, no. 4, pp. 239–248, 2019.
[23] J. Kneller, J. Kalifa, R. Zou et al., “Mechanisms of atrial fibril-
lation termination by pure sodium channel blockade in an
ionically-realistic mathematical model,”Circulation
Research,vol. 96, no. 5, pp. e35–e47, 2005.
[24] M. Liu, K. C. Yang, and S. C. Dudley Jr., “Cardiac sodium
channel mutations: why so many phenotypes?,”Nature
Reviews Cardiology, vol. 11, no. 10, pp. 607–615, 2014.
[25] A. D. Kaplan, H. C. Joca, L. Boyman, and M. Greiser, “Cal-
cium signaling silencing in atrial fibrillation: implications
for atrial sodium homeostasis,”International Journal of
Molecular Sciences, vol. 22, no. 19, article 10513, 2021.
[26] K. Andelova, T. Egan Benova, B. Szeiffova Bacova et al., “Car-
diac connexin-43 hemichannels and pannexin 1 channels:
provocative antiarrhythmic targets,”International Journal
of Molecular Sciences, vol. 22, no. 1, p. 260, 2021.
[27] J. A. Jansen, T. A. van Veen, J. M. de Bakker, and H. V. M. van
Rijen, “Cardiac connexins and impulse propagation,”Journal
of Molecular and Cellular Cardiology, vol. 48, no. 1, pp. 76–
82, 2010.
[28] S. Verheule and S. Kaese, “Connexin diversity in the heart:
insights from transgenic mouse models,”Frontiers in Phar-
macology, vol. 4, p. 81, 2013.
[29] N. J. Severs, A. F. Bruce, E. Dupont, and S. Rothery, “Remod-
elling of gap junctions and connexin expression in diseased
myocardium,”Cardiovascular Research, vol. 80, no. 1,
pp. 9–19, 2008.
[30] M. Firouzi, H. Ramanna, B. Kok et al., “Association of human
connexin 40 gene polymorphisms with atrial vulnerability as
a risk factor for idiopathic atrial fibrillation,”Circulation
Research, vol. 95, no. 4, pp. e29–e33, 2004.
[31] Y. Q. Yang, X. Liu, X. L. Zhang et al., “Novel connexin 40 mis-
sense mutations in patients with familial atrial fibrillation,”
Europace: European pacing, arrhythmias, and cardiac electro-
physiology: journal of the working groups on cardiac pacing,
arrhythmias, and cardiac cellular electrophysiology of the
European Society of Cardiology, vol. 12, no. 10, pp. 1421–
1427, 2010.
[32] S. Herrmann, B. Layh, and A. Ludwig, “Novel insights into
the distribution of cardiac HCN channels: an expression
study in the mouse heart,”Journal of Molecular and Cellular
Cardiology, vol. 51, no. 6, pp. 997–1006, 2011.
[33] Y. Ou, X. L. Niu, and F. X. Ren, “Expression of key ion chan-
nels in the rat cardiac conduction system by laser capture
microdissection and quantitative real-time PCR,”Experimen-
tal Physiology, vol. 95, no. 9, pp. 938–945, 2010.
[34] N. Li, D. Y. Chiang, S. Wang et al., “Ryanodine receptor-
mediated calcium leak drives progressive development of an
atrial fibrillation substrate in a transgenic mouse model,”Cir-
culation, vol. 129, no. 12, pp. 1276–1285, 2014.
[35] D. DiFrancesco, “Funny channels in the control of cardiac
rhythm and mode of action of selective blockers,”Pharmaco-
logical Research, vol. 53, no. 5, pp. 399–406, 2006.
16 Cardiovascular Therapeutics
[36] W. Lee, E. R. Woo, and D. G. Lee, “Effect of apigenin isolated
from Aster yomena against Candida albicans: apigenin-
triggered apoptotic pathway regulated by mitochondrial cal-
cium signaling,”Journal of Ethnopharmacology, vol. 231,
pp. 19–28, 2019.
[37] Z. Y. Xu, Y. Xu, X. F. Xie et al., “Anti-platelet aggregation of
Panax notoginseng triol saponins by regulating GP1BA for
ischemic stroke therapy,”Chinese medicine, vol. 16, no. 1,
p. 12, 2021.
[38] C. Wang, H. Wang, J. H. Xiao, J. L. Wang, J. Z. Xiang,
and Q. Tang, “Inhibitory effects of neferine on Nav 1.5
channels expressed in HEK293 cells,”Journal of Huazhong
University of Science and Technology, vol. 36, no. 4,
pp. 487–493, 2016.
[39] Y. Wang, L. Tu, Y. Li, D. Chen, and S. Wang, “Notoginseno-
side R1 protects against neonatal cerebral hypoxic-ischemic
injury through estrogen receptor-dependent activation of
endoplasmic reticulum stress pathways,”The Journal of
Pharmacology and Experimental Therapeutics, vol. 357,
no. 3, pp. 591–605, 2016.
[40] C. J. Yue and J. J. Zhong, “Manipulation of ginsenoside het-
erogeneity of Panax notoginseng cells in flask and bioreactor
cultivations with addition of phenobarbital,”Bioprocess and
biosystems engineering, vol. 31, no. 2, pp. 95–100, 2008.
[41] K. Chen, L. Liu, X. Zhang et al., “Phased secondary small
interfering RNAs in Panaxnotoginseng,”BMC Genomics,
vol. 19, no. S1, Supplement 1, p. 41, 2018.
[42] C. X. Li, J. Yu, and Y. F. Lu, “Research progress on cardiovas-
cular and cerebrovascular protective effect and mechanism of
tanshinones,”Chinese Journal of New Drugs and Clinical,
vol. 35, no. 8, pp. 542–546, 2016.
[43] C. L. Jia, Y. Chen, and T. Zhang, “Mechanism of notoginse-
noside preventing the development of paroxysmal AF
induced by isoproterenol in mice,”Journal of Integrative
Medicine and Cardio Cerebrovascular Disease, vol. 16, no. 3,
pp. 291–295, 2018.
[44] C. Colson, H. Mittre, A. Busson et al., “Unusual clinical
description of adult with Timothy syndrome, carrier of a
new heterozygote mutation of CACNA1C,”European Journal
of Medical Genetics, vol. 62, no. 7, article 103648, 2019.
[45] H. H. Jiang, Y. Nian, and A. M. Zhang, “Effects of total noto-
ginseng glycosides on voltage dependent calcium and potas-
sium channels,”Chinese Journal of Pharmacology and
Toxicology, vol. 30, no. 9, pp. 921–927, 2016.
[46] K. Dawson, R. Wakili, B. Ordög et al., “MicroRNA29,”Circu-
lation, vol. 127, no. 14, pp. 1466–1475, 2013.
[47] Y. D. Li, Y. F. Hong, Y. Zhang et al., “Association between
reversal in the expression of hyperpolarization-activated
cyclic nucleotide-gated (HCN) channel and age-related atrial
fibrillation,”Medical Science Monitor, vol. 20, pp. 2292–2297,
2014.
[48] Y. Lu, Y. Zhang, N. Wang et al., “MicroRNA-328 contributes
to adverse electrical remodeling in atrial fibrillation,”Circula-
tion, vol. 122, no. 23, pp. 2378–2387, 2010.
[49] Y. J. Chen, J. D. Li, and Q. F. Huang, “Effects of Panax noto-
ginseng saponins on rat cardiomyocytes apoptosis induced
by angiotengin II in vitro,”China Journal of Chinese Materia
Medica, vol. 30, no. 10, pp. 778–781, 2005.
[50] D. D. Wu, C. L. Jia, Y. Chen, and T. Zhang, “Intervention
effects of Panax notoginseng saponins on acetylcholine cal-
cium chloride induced atrial fibrillation and prephase con-
traction in rats,”Shanghai Journal of traditional Chinese
Medicine, vol. 51, no. 3, pp. 70–75, 2017.
[51] N. van den Berg, M. Kawasaki, B. Fabrizi et al., “Epicardial
and endothelial cell activation concurs with extracellular
matrix remodeling in atrial fibrillation,”Clinical and Transla-
tional Medicine, vol. 11, no. 11, article e558, 2021.
[52] R. R. Kuang, L. P. Yuan, X. Z. He et al., “Effects of Panax
notoginseng saponins combined with amiodarone on left
atrial diameter and cardiac anchor repeat protein in paroxys-
mal atrial fibrillation,”Tianjin Traditional Chinese Medicine,
vol. 35, no. 8, pp. 573–575, 2018.
[53] D. Song, J. Hao, and D. Fan, “Biological properties and clini-
cal applications of berberine,”Frontiers of Medicine, vol. 14,
no. 5, pp. 564–582, 2020.
[54] Y. Cai, Q. Xin, J. Lu et al., “A new therapeutic candidate for
cardiovascular diseases: berberine,”Frontiers in Pharmacol-
ogy, vol. 12, article 631100, 2021.
[55] L. R. Wong, E. A. Tan, M. Lim et al., “Functional effects of
berberine in modulating mitochondrial dysfunction and
inflammatory response in the respective amyloidogenic cells
and activated microglial cells - In vitro models simulating
Alzheimer's disease pathology,”Life Sciences, vol. 282, article
119824, 2021.
[56] H. Wu, Q. Chen, J. Liu et al., “Microbiome analysis reveals
gut microbiota alteration in mice with the effect of matrine,”
Microbial pathogenesis, vol. 156, p. 104926, 2021.
[57] T. Joshi, A. K. Singh, P. Haratipour et al., “Targeting AMPK
signaling pathway by natural products for treatment of diabe-
tes mellitus and its complications,”Journal of Cellular Physi-
ology, vol. 234, no. 10, pp. 17212–17231, 2019.
[58] W. H. Ko, X. Q. Yao, C. W. Lau et al., “Vasorelaxant and anti-
proliferative effects of berberine,”European Journal of Phar-
macology, vol. 399, no. 2-3, pp. 187–196, 2000.
[59] K. Qian, C. Y. Tang, L. Y. Chen et al., “Berberine reverses
breast cancer multidrug resistance based on fluorescence
PharmacokineticsIn VitroandIn vivo,”ACS Omega, vol. 6,
no. 16, pp. 10645–10654, 2021.
[60] Z. W. Zhou, H. C. Zheng, L. F. Zhao et al., “Effect of berberine
on acetylcholine-induced atrial fibrillation in rabbit,”Ameri-
can journal of translational research, vol. 7, no. 8, pp. 1450–
1457, 2015.
[61] E. Mirhadi, M. Rezaee, and B. Malaekeh-Nikouei, “Nano
strategies for berberine delivery, a natural alkaloid of _Ber-
beris_,”Biomedicine & Pharmacotherapy = Biomedecine &
pharmacotherapie, vol. 104, pp. 465–473, 2018.
[62] W. M. Huang, Z. H. Zhang, and Y. Q. Xu, “Study of the effects
and mechanisms of berberine on slow-response action poten-
tials,”Journal of Electrocardiology, vol. 23, no. 3, pp. 231–234,
1990.
[63] Y. X. Wang, Y. M. Zheng, and X. B. Zhou, “Inhibitory effects
of berberine on ATP-sensitive K
+
channels in cardiac myo-
cytes,”European Journal of Pharmacology, vol. 316, no. 2-3,
pp. 307–315, 1996.
[64] W. Chang, K. Li, F. Guan et al., “Berberine pretreatment con-
fers cardioprotection against ischemia-reperfusion injury in a
rat model of type 2 diabetes,”Journal of Cardiovascular Phar-
macology and Therapeutics, vol. 21, no. 5, pp. 486–494, 2016.
[65] T. Wei, Z. Liang, Y. Jin, and L. Zhang, “Effects of berberine,
liensinine and neferine on HERG channel expression,”Chi-
nese Journal of Traditional Chinese Medicine, vol. 38, no. 2,
pp. 239–244, 2013.
17Cardiovascular Therapeutics
[66] S. N. Wu, H. S. Yu, C. R. Jan, H. F. Li, and C. L. Yu, “Inhibi-
tory effects of berberine on voltage- and calcium-activated
potassium currents in human myeloma cells,”Life sciences,
vol. 62, no. 25, pp. 2283–2294, 1998.
[67] S. Z. Xu, Y. Zhang, J. Y. Ren, and Z. N. Zhou, “Effects of ber-
berine of L- and T-type calcium channels in guinea pig ven-
tricular myocytes,”Acta pharmacologica Sinica, vol. 18,
no. 6, pp. 515–518, 1997.
[68] M. Y. Qi, Y. Feng, D. Z. Dai, N. Li, Y. S. Cheng, and Y. Dai,
“CPU86017, a berberine derivative, attenuates cardiac failure
through normalizing calcium leakage and downregulated
phospholamban and exerting antioxidant activity,”Acta
Pharmacologica Sinica, vol. 31, no. 2, pp. 165–174, 2010.
[69] H. L. Wang, D. Z. Dai, E. Gao, Y. P. Zhang, and F. Lu, “Dis-
persion of ventricular mRNA of RyR2 and SERCA2 associ-
ated with arrhythmogenesis in rats,”Acta pharmacologica
Sinica, vol. 25, no. 6, pp. 738–743, 2004.
[70] H. Chen, Y. Chen, Y. Tang et al., “Berberine attenuates spon-
taneous action potentials in sinoatrial node cells and the cur-
rents of human HCN4 channels expressed in Xenopus laevis
oocytes,”Molecular Medicine Reports, vol. 10, no. 3,
pp. 1576–1582, 2014.
[71] H. Zheng, F. Zhu, P. Miao, Z. Mao, D. P. Redfearn, and R. Y.
Cao, “Antimicrobial natural product berberine is efficacious
for the treatment of atrial fibrillation,”Bio Med research
international, vol. 2017, article 3146791, pp. 1–5, 2017.
[72] D. F. Shen, Q. Z. Tang, L. Yan et al., “Tetrandrine blocks
cardiac hypertrophy by disrupting reactive oxygen species-
dependent ERK1/2 signalling,”British Journal of Pharma-
cology, vol. 159, no. 4, pp. 970–981, 2010.
[73] X. C. Yu, S. Wu, C. F. Chen, K. T. Pang, and T. M. Wong,
“Antihypertensive and anti-arrhythmic effects of an extract
of Radix Stephaniae Tetrandrae in the rat,”The Journal of
pharmacy and pharmacology, vol. 56, no. 1, pp. 115–122,
2004.
[74] M. Müller, S. Gerndt, Y. K. Chao et al., “Gene editing and
synthetically accessible inhibitors reveal role for TPC2 in
HCC cell proliferation and tumor growth,”Cell chemical biol-
ogy,vol. 28, no. 8, pp. 1119–1131.e27, 2021.
[75] R. Wang, T. M. Ma, F. Liu, and H. Q. Gao, “Research progress
on pharmacological action and clinical application of Stepha-
nia tetrandrae Radix,”China Journal of Chinese Materia
Medica, vol. 42, no. 4, pp. 634–639, 2017.
[76] T. J. Zhang, R. X. Guo, X. Li, Y. W. Wang, and Y. J. Li, “Tet-
randrine cardioprotection in ischemia-reperfusion (I/R)
injury via JAK3/STAT3/Hexokinase II,”European journal
of pharmacology, vol. 813, pp. 153–160, 2017.
[77] Q. Li, L. Chang, D. M. Su, and X. Ma, “Effects of tetrandrine
on proliferation and activation of cardiac fibroblasts,”Health
Sciences, vol. 50, no. 2, pp. 331–334, 2018.
[78] I. Huber, E. Wappl, A. Herzog et al., “Conserved Ca2+-antag-
onist-binding properties and putative folding structure of a
recombinant high-affinity dihydropyridine-binding
domain,”The Biochemical Journal, vol. 347, no. 3, Part 3,
pp. 829–836, 2000.
[79] S. N. Wu, H. F. Li, and Y. C. Lo, “Characterization of
tetrandrine-induced inhibition of large-conductance
calcium-activated potassium channels in a human endothe-
lial cell line (HUV-EC-C),”The Journal of pharmacology
and experimental therapeutics, vol. 292, no. 1, pp. 188–195,
2000.
[80] N. Bhagya and K. R. Chandrashekar, “Tetrandrine - A mole-
cule of wide bioactivity,”Phytochemistry, vol. 125, pp. 5–13,
2016.
[81] H. X. Wang, C. Y. Kwan, and T. M. Wong, “Tetrandrine
inhibits electrically induced [Ca
2+
]
i
transient in the isolated
single rat cardiomyocyte,”European Journal of Pharmacol-
ogy, vol. 319, no. 1, pp. 115–122, 1997.
[82] M. R. Rao, “Effects of tetrandrine on cardiac and vascular
remodeling,”Acta pharmacologica Sinica, vol. 23, no. 12,
pp. 1075–1085, 2002.
[83] L. S. Rubio, G. Garrido, L. Llanes, and J. L. Alvarez, “Effects of
Tetrandrine on Ca
2+
- and Na
+
-Currents of Single Bullfrog
Cardiomyocytes,”Journal of Molecular and Cellular Cardiol-
ogy, vol. 25, no. 7, pp. 801–813, 1993.
[84] L. Chen, Q. Y. Li, Y. Yang, Z. W. Li, and X. R. Zeng, “Inhib-
itory effects of tetrandrine on the Na
+
channel of human
atrial fibrillation myocardium,”Acta Pharmacologica Sinica,
vol. 30, no. 2, pp. 166–174, 2009.
[85] J. S. Xia, D. L. Guo, Y. Zhang, Z. N. Zhou, F. D. Zeng, and C. J.
Hu, “Inhibitory effects of dauricine on potassium currents in
guinea pig ventricular myocytes,”Acta pharmacologica
Sinica, vol. 21, no. 1, pp. 60–64, 2000.
[86] L. Li, L. Ma, D. Wang et al., “Design and synthesis of matrine
derivatives as novel anti-pulmonary fibrotic agents via
repression of the TGFβ/Smad pathway,”Molecules, vol. 24,
no. 6, p. 1108, 2019.
[87] J. Shi, G. Han, J. Wang et al., “Matrine promotes hepatic oval
cells differentiation into hepatocytes and alleviates liver
injury by suppression of Notch signalling pathway,”Life Sci-
ences, vol. 261, article 118354, 2020.
[88] M. Dai, N. Chen, J. Li et al., “In vitro and in vivo anti-
metastatic effect of the alkaliod matrine from _Sophora flave-
cens_ on hepatocellular carcinoma and its mechanisms,”
Phytomedicine, vol. 87, article 153580, 2021.
[89] C. Y. Woo, J. Y. Baek, A. R. Kim et al., “Inhibition of ceramide
accumulation in podocytes by myriocin prevents diabetic
nephropathy,”Diabetes & Metabolism Journal, vol. 44,
no. 4, pp. 581–591, 2020.
[90] C. J. Dickson, C. Velez-Vega, and J. S. Duca, “Revealing
molecular determinants of hERG blocker and activator bind-
ing,”Journal of Chemical Information and Modeling, vol. 60,
no. 1, pp. 192–203, 2020.
[91] S. Saponara, F. Fusi, D. Iovinelli et al., “Flavonoids and hERG
channels: friends or foes?,”European Journal of Pharmacol-
ogy, vol. 899, article 174030, 2021.
[92] S. L. Wilson, C. E. Dempsey, J. C. Hancox, and N. V. Marrion,
“Identification of a proton sensor that regulates conductance
and open time of single hERG channels,”Scientific Reports,
vol. 9, no. 1, p. 19825, 2019.
[93] B. Hille, “Gprotein-coupled mechanisms and nervous signal-
ing,”Neuron,vol. 9, no. 2, pp. 187–195, 1992.
[94] X. H. Sui, Y. H. Zhou, Z. Q. Li et al., “The effects of matrine on
IKM3 atrial myo⁃cytes of canine with atrial fibrillation,”Chi-
nese Pharmacological Bulletin, vol. 24, no. 2, pp. 199–202,
2008.
[95] J. Zhou, W. Ma, X. Wang et al., “Matrine suppresses reactive
oxygen species (ROS)-mediated MKKs/p38-Induced inflam-
mation in oxidized low-density lipoprotein (ox-LDL)-stimu-
lated macrophages,”Medical science monitor: international
medical journal of experimental and clinical research,
vol. 25, pp. 4130–4136, 2019.
18 Cardiovascular Therapeutics
[96] Y. Zhou, W. Xu, R. Han et al., “Matrine inhibits pacing
induced atrial fibrillation by modulating I (KM3) and I (Ca-
L),”International journal of biological sciences, vol. 8, no. 1,
pp. 150–158, 2012.
[97] L. Reilly and L. L. Eckhardt, “Cardiac potassium inward rec-
tifier Kir2: Review of structure, regulation, pharmacology,
and arrhythmogenesis,”Heart Rhythm, vol. 18, no. 8,
pp. 1423–1434, 2021.
[98] B. Huang, D. Qin, and N. El-Sherif, “Spatial alterations of Kv
channels expression and K(+) currents in post-MI remodeled
rat heart,”Cardiovascular Research, vol. 52, no. 2, pp. 246–
254, 2001.
[99] X. Peng, L. Lin, X. Q. Zhou, D. Y. Yang, and T. Y. Yin, “Effect
of matrine on helper T cell 1/helper T cell 2 balance and
potassium channels Kv2.1 and Kir 2.1 in rats with myocardial
infarction,”Journal of Clinical Medicine, vol. 37, no. 2,
pp. 121–124, 2020.
[100] S. T. Hu, J. Ba, and H. M. Liu, “Abnormal calcium induced
calcium release in cardiomyocytes of rats with chronic heart
failure,”Ningxia Medical Journal, vol. 35, no. 12, pp. 1131–
1133, 2013.
[101] J. Ma, S. Ma, C. Yin, and H. Wu, “Matrine reduces suscepti-
bility to postinfarct atrial fibrillation in rats due to antifibrotic
properties,”Journal of Cardiovascular Electrophysiology,
vol. 29, no. 4, pp. 616–627, 2018.
[102] M. L. Li, H. Q. Tang, Y. Wang et al., “Effects of matrine
on action potential and sodium channel of guinea pig ven-
tricular myocytes,”Chinese Journal of Basic Medicine of
Traditional Chinese Medicine, vol. 18, no. 11, pp. 1219–
1221, 2012.
[103] H. Q. Tang, M. L. Li, Y. Wang, J. Guo, Y. Z. Pang, and G. Z.
Li, “Experimental study on the effect of matrine on action
potential of guinea pig papillary muscle cells,”Chinese Jour-
nal of Experimental Formulary, vol. 18, no. 24, pp. 241–243,
2012.
[104] X. Jin, Y. Jiang, G. Xue et al., “Increase of late sodium current
contributes to enhanced susceptibility to atrial fibrillation in
diabetic mice,”European Journal of Pharmacology, vol. 857,
article 172444, 2019.
[105] H. J. Park, M. Gholam Zadeh, J. H. Suh, and H. S. Choi,
“Dauricine protects from LPS-induced bone loss via the
ROS/PP2A/NF-κB axis in osteoclasts,”Antioxidants, vol. 9,
no. 7, p. 588, 2020.
[106] L. Jiang, T. Guo, Y. Jiang, P. Liu, and Y. Bai, “Dauricine
inhibits human pancreatic carcinoma cell proliferation
through regulating miRNAs,”Molecular Omics, vol. 17,
no. 4, pp. 630–640, 2021.
[107] T. Guo, Y. Z. Zhang, D. X. Liu, P. Zou, and D. Shen, “Daur-
icine inhibits redistribution of platelet membrane glycopro-
tein IV and release of intracellular alpha-granule
thrombospondin induced by thrombin,”Acta pharmacolo-
gica Sinica, vol. 20, no. 6, pp. 533–536, 1999.
[108] X. F. Yan, M. Ding, and L. L. Zhou, “Mechanoelectric feed-
back effects induced by increased atrial pressure in isolated
rabbits treated with batophenanthroline,”Chinese Journal of
emergency resuscitation and disaster medicine, vol. 4, no. 3,
pp. 142–144, 2007.
[109] J. Zhao, Y. Lian, C. Lu, L. Jing, H. Yuan, and S. Peng, “Inhib-
itory effects of a bisbenzylisoquinline alkaloid dauricine on
HERG potassium channels,”Journal of ethnopharmacology,
vol. 141, no. 2, pp. 685–691, 2012.
[110] J. Q. Qian, “Cardiovascular pharmacological effects of bis-
benzylisoquinoline alkaloid derivatives,”Acta pharmacolo-
gica Sinica, vol. 23, no. 12, pp. 1086–1092, 2002.
[111] Q. N. Liu, X. Y. Yang, L. Zhang et al., “Therapeutic effect of
Menispermum dauricum on early and late afterdepolariza-
tions induced by dofetilide,”Medical Reports, vol. 4, no. 1,
pp. 15–18, 2008.
[112] Q. N. Liu, L. Zhang, P. L. Gong, X. Y. Yang, and F. D. Zeng,
“Inhibitory effects of dauricine on early afterdepolarizations
and L-type calcium current,”Canadian Journal Of Physiology
and Pharmacology,vol. 87, no. 11, pp. 954–962, 2009.
[113] L. Xia, Protective effect of dauricine on hypertrophic myocar-
dium and its mechanism, Master's thesis of Huazhong Uni-
versity of science and technology, 2006.
[114] Y. Q. Yang, X. L. Zhang, X. H. Wang et al., “Connexin 40
nonsense mutation in familial atrial fibrillation,”Interna-
tional journal of molecular medicine, vol. 26, no. 4, pp. 605–
610, 2010.
[115] J. M. Zhang, D. Q. Li, Y. B. Feng, S. L. Yu, and H. Z. Hu,
“Batophenanthroline attenuates gap junction protein 40 deg-
radation during rapid atrial pacing in rabbits,”Chinese Jour-
nal of pathophysiology, vol. 4, no. 4, p. 173, 2004.
[116] N. S. Narayana Moorthy, M. J. Ramos, and P. A. Fernandes,
“Human ether-a-go-go-related gene channel blockers and
its structural analysis for drug design,”Current Drug Targets,
vol. 14, no. 1, pp. 102–113, 2013.
[117] L. Zou, K. Yu, Y. Fan et al., “The inhibition by Guanfu base A
of neuropathic pain mediated by P2Y12Receptor in dorsal
root ganglia,”ACS chemical neuroscience, vol. 10, no. 3,
pp. 1318–1325, 2019.
[118] W. Liu, X. Wang, X. Zhou, H. Duan, P. Zhao, and W. Liu,
“Quantitative structure-activity relationship between the tox-
icity of amine surfactant and its molecular structure,”The Sci-
ence of the Total Environment, vol. 702, article 134593, 2020.
[119] M. Wang, J. Zhu, Y. M. Yang et al., “Effect of Guanfu a hydro-
chloride on canine vagal atrial fibrillation model,”Chinese
Journal of Cardiac Pacing and Electrophysiology, vol. 4,
no. 2, pp. 148–152, 2008.
[120] F. Xiong, K. Liu, S. Liu et al., “Safety, heart specificity, and
therapeutic effect evaluation of Guanfu base A-loaded solid
nanolipids in treating arrhythmia,”Drug delivery and trans-
lational research, vol. 8, no. 5, pp. 1471–1482, 2018.
[121] J. Sun, Y. Peng, H. Wu et al., “Guanfu base A, an antiarrhyth-
mic alkaloid of Aconitum coreanum, is a CYP2D6 inhibitor
of human, monkey, and dog isoforms,”Drug Metabolism
and Disposition, vol. 43, no. 5, pp. 713–724, 2015.
[122] B. Bortoleti, M. D. Gonçalves, F. Tomiotto-Pellissier et al.,
“Grandiflorenic acid promotes death of promastigotes via
apoptosis-like mechanism and affects amastigotes by increas-
ing total iron bound capacity,”Phytomedicine, vol. 46,
pp. 11–20, 2018.
[123] X. F. Huang, Y. M. Yang, J. Zhu, Y. Dai, and J. L. Pu, “Effect of
kaempferol hydrochloride on F656C mutant potassium
channels in the HERG gene,”China Journal of Chinese Mate-
ria Medica, vol. 19, no. 7, pp. 612–616, 2010.
[124] Z. L. Qi, Z. Wang, W. Li et al., “Nephroprotective effects of
anthocyanin from the fruits of Panax ginseng (GFA) on
cisplatin-induced acute kidney injury in mice,”Phytotherapy
Research, vol. 31, no. 9, pp. 1400–1409, 2017.
[125] M. K. Rowe and J. D. Roberts, “The evolution of gene-guided
management of inherited arrhythmia syndromes: peering
19Cardiovascular Therapeutics
beyond monogenic paradigms towards comprehensive geno-
mic risk scores,”Journal of Cardiovascular Electrophysiology,
vol. 31, no. 11, pp. 2998–3008, 2020.
[126] X. Gao, J. L. Pu, Y. M. Yang et al., “Effects of Guanfu a hydro-
chloride on L-type calcium channels in rat ventricular myo-
cyte membrane (Ca-L),”Chinese Journal of New Drugs,
vol. 4, no. 22, pp. 1926–1929, 2006.
[127] J. Gao, T. Wang, X. Yao et al., “Clinical evidence-guided net-
work pharmacology analysis reveals a critical contribution of
β1-adrenoreceptor upregulation to bradycardia alleviation by
Shenxian-Shengmai,”BMC Complementary and Alternative
Medicine, vol. 19, no. 1, p. 357, 2019.
[128] Y. Chu, Q. Yang, L. Ren et al., “Late sodium current in atrial
cardiomyocytes contributes to the induced and spontaneous
atrial fibrillation in rabbit hearts,”Journal of Cardiovascular
Pharmacology, vol. 76, no. 4, pp. 437–444, 2020.
[129] J. C. Makielski, “Late sodium current: a mechanism for
angina, heart failure, and arrhythmia,”Trends in Cardiovas-
cular Medicine, vol. 26, no. 2, pp. 115–122, 2016.
[130] L. Belardinelli, J. C. Shryock, and H. Fraser, “Inhibition of the
late sodium current as a potential cardioprotective principle:
effects of the late sodium current inhibitor ranolazine,”Heart,
vol. 92, Supplement 4, pp. iv6–iv14, 2006.
[131] X. H. Shao, Evaluation of Guanfu a hydrochloride on late
sodium current in SCN5A gene mutation, [Ph.D. thesis],
Peking Union Medical College, 2014.
[132] J. Lin, M. Wu, and J. W. Zhang, “Guanfacine hydrochloride
terminates paroxysmal atrioventricular reentrant tachycardia
clinical study,”Yeungnan Journal of cardiovascular disease,
vol. 17, no. S1, p. 110, 2011.
[133] P. Wicha, A. Onsa-Ard, W. Chaichompoo, A. Suksamrarn,
and C. Tocharus, “Vasorelaxant and antihypertensive effects
of neferine in rats: an in vitro and in vivo study,”Planta Med-
ica, vol. 86, no. 7, pp. 496–504, 2020.
[134] L. Yang, Y. Xiong, Z. Sun, X. Lin, and H. Ni, “Neferine
inhibits 7, 12-dimethylbenz (a)anthracene-induced mam-
mary tumorigenesis by suppression of cell proliferation and
induction of apoptosis via modulation of the PI3K/AKT/
NF-κB signaling pathway,”Journal of environmental pathol-
ogy, toxicology and oncology: official organ of the Interna-
tional Society for Environmental Toxicology and Cancer,
vol. 40, no. 3, pp. 51–61, 2021.
[135] Z. Qi, R. Wang, R. Liao, S. Xue, and Y. Wang, “Neferine ame-
liorates sepsis-induced myocardial dysfunction through anti-
apoptotic and antioxidative effects by regulating the PI3K/
AKT/mTOR signaling pathway,”Frontiers in Pharmacology,
vol. 12, article 706251, 2021.
[136] R. P. Yang, Y. J. Zhou, W. Song, Z. Yin, A. D. He, and Z. Y.
Ming, “Pharmacological actions of neferine in the modula-
tion of human platelet function,”European journal of phar-
macology, vol. 862, p. 172626, 2019.
[137] K. M. Chiu, Y. L. Hung, S. J. Wang et al., “Anti-allergic and
anti-inflammatory effects of neferine on RBL-2H3 cells,”
International Journal of Molecular Sciences, vol. 22, no. 20,
article 10994, 2021.
[138] T. Xu, D. Singh, J. Liu et al., “Neferine, is not inducer but
blocker for macroautophagic flux targeting on lysosome mal-
function,”Biochemical and biophysical research communica-
tions, vol. 495, no. 1, pp. 1516–1521, 2018.
[139] Z. X. Dong, X. Zhao, D. F. Gu et al., “Comparative effects of
liensinine and neferine on the human ether-a-go-go-related
gene potassium channel and pharmacological activity analy-
sis,”Cellular Physiology and Biochemistry, vol. 29, no. 3-4,
pp. 431–442, 2012.
[140] Z. Zuo, L. He, X. Duan, Z. Peng, and J. Han, “Glycyrrhizic
acid exhibits strong anticancer activity in colorectal cancer
cells via SIRT3 inhibition,”Bioengineered, 2021.
[141] J. Yu and W. S. Hu, “Effects of neferine on platelet aggrega-
tion in rabbits,”Acta Pharmaceutica Sinica, vol. 32, no. 1,
pp. 1–4, 1997.
[142] J. Wan, L. Zhao, C. Xu et al., “Effects of neferine on the phar-
macokinetics of amiodarone in rats,”Biomedical Chromatog-
raphy: BMC, vol. 25, no. 8, pp. 858–866, 2011.
[143] H. Shan, X. Li, Z. Pan et al., “Tanshinone IIA protects against
sudden cardiac death induced by lethal arrhythmias via
repression of microRNA-1,”British Journal of Pharmacology,
vol. 158, no. 5, pp. 1227–1235, 2009.
[144] Y. Fang, C. Duan, S. Chen et al., “Tanshinone-IIA inhibits
myocardial infarct via decreasing of the mitochondrial apo-
ptotic signaling pathway in myocardiocytes,”International
Journal of Molecular Medicine, vol. 48, no. 2, p. 158, 2021.
[145] Y. Wang, W. Jin, and J. Wang, “Tanshinone IIA regulates
microRNA‑125b/foxp3/caspase‑1 signaling and inhibits cell
viability of nasopharyngeal carcinoma,”Molecular Medicine
Reports, vol. 23, no. 5, p. 371, 2021.
[146] J. Wen, Y. Chang, S. Huo et al., “Tanshinone IIA attenuates
atherosclerosis via inhibiting NLRP3 inflammasome activa-
tion,”Aging, vol. 13, no. 1, pp. 910–932, 2020.
[147] J. Li, Y. Chen, L. Zhang et al., “Total saponins of panaxnoto-
ginseng promotes lymphangiogenesis by activation VEGF-C
expression of lymphatic endothelial cells,”Journal of Ethno-
pharmacology, vol. 193, pp. 293–302, 2016.
[148] X. Liu, Y. Li, H. Zhang, Y. Ji, Z. Zhao, and C. Wang, “The
research of ion channel-related gene polymorphisms with
atrial fibrillation in the Chinese Han population,”Molecular
Genetics & Genomic Medicine, vol. 7, no. 8, article e835,
2019.
[149] S. Mao, S. Taylor, Q. Chen, M. Zhang, and A. Hinek, “Sodium
tanshinone IIA sulfonate prevents the adverse left ventricular
remodelling: focus on polymorphonuclear neutrophil-
derived granule components,”Journal of Cellular and Molec-
ular Medicine, vol. 23, no. 7, pp. 4592–4600, 2019.
[150] Z. M. Li, S. W. Xu, and P. Q. Liu, “_Salvia miltiorrhiza_ Burge
(Danshen): a golden herbal medicine in cardiovascular ther-
apeutics,”Acta Pharmacologica Sinica, vol. 39, no. 5,
pp. 802–824, 2018.
[151] R. Yang, D. D. Zhao, and X. L. Li, “Research progress of drug
targeting micro RNA for cardiovascular disease,”Chinese
Journal of clinical pharmacology, vol. 32, no. 8, pp. 762–764,
2016.
[152] L. Yang, X. J. Zou, Z. Yi, and H. Z. Hao, “Effects of sodium
tanshinone II_A sulfonate on collagen synthesis and TGF-
β_1 activation in atrial fibroblasts induced by Ang II,”Chi-
nese journal of traditional Chinese medicine, vol. 39, no. 6,
pp. 1093–1096, 2014.
[153] T. Chen, M. Li, X. Fan, J. Cheng, and L. Wang, “Sodium tan-
shinone IIA sulfonate prevents angiotensin II-induced differ-
entiation of human atrial fibroblasts into myofibroblasts,”
Oxidative Medicine and Cellular Longevity, vol. 2018, Article
ID 6712585, 10 pages, 2018.
[154] Y. Y. Wang and G. X. Xie, “Meta analysis of tanshinone IIA-
sulfonate combined with amiodarone in the treatment of
20 Cardiovascular Therapeutics
acute myocardial infarction with atrial fibrillation,”World
Journal of Traditional Chinese Medicine, vol. 16, no. 5, 2010.
[155] J. Hernandez-Cascales, “Resveratrol enhances the inotropic
effect but inhibits the proarrhythmic effect of sympathomi-
metic agents in rat myocardium,”Peer J, vol. 5, no. e,
p. e3113, 2017.
[156] M. H. Hsu, K. A. Chang, Y. C. Chen, I. Lin, J. M. Sheen, and
L. T. Huang, “Resveratrol prevented spatial deficits and res-
cued disarrayed hippocampus asymmetric dimethylarginine
and brain-derived neurotrophic factor levels in young rats
with increased circulating asymmetric dimethylarginine,”
NeuroReport, vol. 32, no. 13, pp. 1091–1099, 2021.
[157] T. C. Almeida, G. N. da Silva, D. V. de Souza et al., “Resver-
atrol effects in oral cancer cells: a comprehensive review,”
Medical Oncology, vol. 38, no. 8, p. 97, 2021.
[158] H. Kazemirad and H. R. Kazerani, “Cardioprotective effects
of resveratrol following myocardial ischemia and reperfu-
sion,”Molecular Biology Reports, vol. 47, no. 8, pp. 5843–
5850, 2020.
[159] Y. Zhang, Y. Lu, M. J. Ong’achwa et al., “Resveratrol inhibits
the TGF-β1-induced proliferation of cardiac fibroblasts and
collagen secretion by downregulating mi R-17 in rat,”Bio
Med research international, vol. 2018, article 8730593, 2018.
[160] S. Barangi, A. W. Hayes, and G. Karimi, “The more effective
treatment of atrial fibrillation applying the natural com-
pounds; as NADPH oxidase and ion channel inhibitors,”
Critical Reviews in Food Science and Nutrition, vol. 58,
no. 7, pp. 1230–1241, 2018.
[161] M. C. Pan, X. W. Zhou, Y. Liu et al., “Research progress on
the molecular mechanisms of toxicology of ethanol-
aconitine induced arrhythmia,”Journal of Forensic Sciences,
vol. 36, no. 1, pp. 115–119, 2020.
[162] W. Liu, P. Chen, J. Deng, J. Lv, and J. Liu, “Resveratrol and
polydatin as modulators of Ca2+ mobilization in the cardio-
vascular system,”Annals of the New York Academy of Sci-
ences, vol. 1403, no. 1, pp. 82–91, 2017.
[163] X. Y. Huang, “Effect of resveratrol on Kv2.1 potassium chan-
nel in H9c2 cardiomyocytes,”Chinese Journal of Cardiac Pac-
ing and Electrophysiology, vol. 34, no. 5, pp. 484–487, 2020.
[164] Y. R. Chen, F. F. Yi, X. Y. Li et al., “Resveratrol attenuates ven-
tricular arrhythmias and improves the long-term survival in
rats with myocardial infarction,”Cardiovascular Drugs and
Therapy, vol. 22, no. 6, pp. 479–485, 2008.
[165] H. Sutanto, D. Dobrev, and J. Heijman, “Resveratrol: an effec-
tive pharmacological agent to prevent inflammation-induced
atrial fibrillation?,”Naunyn-Schmiedeberg's Archives of Phar-
macology, vol. 391, no. 11, pp. 1163–1167, 2018.
[166] C. Qian, J. Ma, P. Zhang et al., “Resveratrol attenuates the
Na
+
-dependent intracellular Ca
2+
overload by inhibiting
H
2
O
2
-induced increase in late sodium current in ventricular
myocytes,”PLoS One, vol. 7, no. 12, article e51358, 2012.
[167] J. X. Du, Y. Li, J. G. Wang et al., “Effects of resveratrol and tri-
methylresveratrol on sodium current in guinea pig ventricu-
lar myocytes,”Journal of Henan University of science and
Technology, vol. 28, no. 3, pp. 161–164, 2010.
[168] C. P. Qian, Resveratrol Inhibits the Increase of Late Sodium
Current Induced by Hydrogen Peroxide in Ventricular Myo-
cytes to Relieve Sodium Dependent Calcium Overload, Wuhan
University of Science and Technology, 2012.
[169] Y. Zhang, S. Zhang, Z. Liu et al., “Resveratrol prevents atrial
fibrillation by inhibiting atrial structural and metabolic
remodeling in collagen-induced arthritis rats,”Naunyn-
Schmiedeberg's archives of pharmacology, vol. 391, no. 11,
pp. 1179–1190, 2018.
[170] J. Song, J. Wang, B. H. Li, D. Wang, D. C. Meng, and Y. Z. Lu,
“A study on anti-arrhythmia mechanisms of resveratrol on
ischemia/reperfusion in rats by regulating PI3K/Akt signal-
ing pathway,”Chinese Journal of Applied Physiology, vol. 33,
no. 3, pp. 239–243, 2017.
[171] X. F. Zhou, Z. R. Wang, Z. Q. Zhang et al., “Effect of resvera-
trol on the expression of deacetylase 1 in chronic atrial fibril-
lation in pigs,”Journal of xuzhou medical college, vol. 30,
no. 5, pp. 318–320, 2010.
[172] Y. Wei, Effects of Resveratrol Glycosides on Calcium and
Potassium Channels in Rat Ventricular Myocytes, [Ph.D. the-
sis], Hebei Medical University, 2012.
[173] I. Baczko, D. Liknes, W. Yang et al., “Characterization of a
novel multifunctional resveratrol derivative for the treatment
of atrial fibrillation,”British Journal of Pharmacology,
vol. 171, no. 1, pp. 92–106, 2014.
[174] G. L. Liu, W. Yang, Y. X. Wang et al., “Effect of puerarin on
arrhythmia induced by myocardial ischemia-reperfusion in
rats and its mechanism,”Genomics and Applied Biology,
vol. 39, no. 7, pp. 3350–3354, 2020.
[175] W. Shuyong, “Research Progress on cardiovascular protec-
tive effect and mechanism of Puerarin,”Chinese Journal of
traditional Chinese Medicine,vol. 40, no. 12, pp. 2278–
2284, 2015.
[176] B. X. Chen, Y. N. Liu, H. Y. Li et al., “Neuroprotective effects
of puerarin on ischemic brain injury in mice through antiox-
idant stress,”Chinese Journal of Clinical Pharmacology,
vol. 37, no. 11, 2010.
[177] Y. Hu, H. Li, R. Li, Z. Wu, W. Yang, and W. Qu, “Puer-
arin protects vascular smooth muscle cells from oxidized
low-density lipoprotein-induced reductions in viability via
inhibition of the p 38 MAPK and JNK signaling path-
ways,”Experimental and Therapeutic Medicine, vol. 20,
no. 6, p. 270, 2020.
[178] G. Yuan, S. Shi, Q. Jia et al., “Use of network pharmacology to
explore the mechanism of Gegen (Puerariae lobatae Radix) in
the treatment of type 2 diabetes mellitus associated with
hyperlipidemia,”Evidence-based complementary and alterna-
tive medicine: eCAM, vol. 2021, article 6633402, 2021.
[179] J. F. Gao, M. M. Liu, Z. P. Guo, C. Hu, Z. Feng, and J. Yan,
“Puerarin alleviated hepatic insulin resistance in type 2 dia-
betic mice through fetuin B-AMPK/ACC signaling pathway,”
Journal of Southern Medical University, vol. 41, no. 6,
pp. 839–846, 2021.
[180] M. Li, L. S. Zhang, J. R. Zhou et al., “Effects of artemisinin and
amiodarone on arrhythmia in rats,”Chongqing Medical,
vol. 49, no. 6, pp. 883–886, 2020.
[181] C. Y. Li, G. T. Zhang, C. Han et al., “Progress in the study of
the molecular mechanism of the antitumor action of robi-
nin,”Chinese Journal of Traditional Chinese Medicine,
vol. 39, no. 5, pp. 53–56, 2021.
[182] E. P. Scholz, E. Zitron, H. A. Katus, and C. A. Karle, “Cardio-
vascular ion channels as a molecular target of flavonoids,”
Cardiovascular Therapeutics, vol. 28, no. 4, pp. e46–e52,
2010.
[183] J. R. Zeng, Q. L. Zeng, Z. H. Huang et al., “Experimental study
of puerarin on isolated atria of guinea pig,”Journal of gannan
medical college, vol. 31, no. 4, pp. 513-514, 2011.
21Cardiovascular Therapeutics
[184] H. Xu, L. Zhang, Z. Gao et al., “Effect of puerarin on action
potential of rat ventricular myocytes,”Advances in modern
biomedicine, vol. 14, no. 19, p. 3635, 2014.
[185] H. Zhang, L. Zhang, Q. Zhang et al., “Puerarin: a novel antag-
onist to inward rectifier potassium channel (IK1),”Molecular
and cellular biochemistry, vol. 352, no. 1-2, pp. 117–123,
2011.
[186] H. Xu, M. Zhao, S. Liang et al., “The effects of puerarin on rat
ventricular myocytes and the potential mechanism,”Scien-
tific reports, vol. 6, no. 1, p. 35475, 2016.
[187] Q. Gao, B. Yang, Z. G. Ye, J. Wang, I. C. Bruce, and Q. Xia,
“Opening the calcium-activated potassium channel partici-
pates in the cardioprotective effect of puerarin,”European
Journal of Pharmacology, vol. 574, no. 2-3, pp. 179–184, 2007.
[188] X. G. Guo, J. Z. Chen, X. Zhang, and Q. Xia, “Effect of puer-
arin on L-type calcium channel in isolated rat ventricular
myocytes,”China Journal of Chinese Materia Medica,
vol. 29, no. 3, pp. 248–251, 2004.
[189] Y. F. Yuan, X. Y. Hu, and X. H. Ou, “Comparison of anti-
arrhythmic effects of puerarin sodium sulfonate and puer-
arin,”Chinese journal of hospital pharmacy, vol. 31, no. 3,
pp. 196–198, 2011.
[190] L. Zhang, H. X. Wang, J. H. Gao et al., “Effect of puerarin on
myocardial collagen and intracellular calcium in rat cardiac
hypertrophy induced by isoproterenol,”Proprietary Chinese
medicine, vol. 8, pp. 1220–1222, 2008.
[191] G. Q. Zhang, X. M. Hao, D. Z. Dai, Y. Fu, P. A. Zhou, and
C. H. Wu, “Puerarin blocks Na+ current in rat ventricular
myocytes,”Acta pharmacologica Sinica, vol. 24, no. 12,
pp. 1212–1216, 2003.
[192] M. P. Wan, “Puerarin combined with amidaphone in the
treatment of atrial fibrillation (87 cases),”Aerospace Medi-
cine, vol. 20, no. 12, pp. 17-18, 2009.
[193] G. R. Li, H. B. Wang, G. W. Qin et al., “Acacetin, a natural fla-
vone, selectively inhibits human atrial repolarization potas-
sium currents and prevents atrial fibrillation in dogs,”
Circulation, vol. 117, no. 19, pp. 2449–2457, 2008.
[194] J. Li, D. Xu, L. Wang et al., “Glycyrrhizic acid inhibits SARS-
CoV-2 infection by blocking spike protein-mediated cell
attachment,”Molecules, vol. 26, no. 20, p. 6090, 2021.
[195] G. Zhang, Z. Li, J. Dong et al., “Acacetin inhibits invasion,
migration and TGF-β1-induced EMT of gastric cancer cells
through the PI3K/Akt/Snail pathway,”BMC complementary
medicine and therapies, vol. 22, no. 1, p. 10, 2022.
[196] S. Xie, Y. Zhang, L. Xu et al., “Acacetin attenuates Streptococ-
cus suis virulence by simultaneously targeting suilysin and
inflammation,”Microbial pathogenesis, vol. 162, p. 105354,
2021.
[197] Y. Wei, P. Yuan, Q. Zhang et al., “Acacetin improves endo-
thelial dysfunction and aortic fibrosis in insulin-resistant
SHR rats by estrogen receptors,”Molecular Biology Reports,
vol. 47, no. 9, pp. 6899–6918, 2020.
[198] C. Jalili, N. Akhshi, F. Raissi et al., “Acacetin alleviates hepa-
titis following renal ischemia-reperfusion in male Balb/C
mice by antioxidants regulation and inflammatory markers
suppression,”Journal of Investigative Surgery, vol. 34, no. 5,
pp. 495–503, 2021.
[199] H. Liu, Y. J. Wang, L. Yang et al., “Synthesis of a highly water-
soluble acacetin prodrug for treating experimental atrial
fibrillation in beagle dogs,”Scientific Reports, vol. 6, no. 1,
article 25743, 2016.
[200] J. M. Di Diego, B. Patocskai, H. Barajas-Martinez et al., “Aca-
cetin suppresses the electrocardiographic and arrhythmic
manifestations of the J wave syndromes,”PLoS One, vol. 15,
no. 11, article e0242747, 2020.
[201] H. J. Wu, W. Wu, H. Y. Sun et al., “Acacetin causes a fre-
quency- and use-dependent blockade of hKv1.5 channels by
binding to the S6 domain,”Journal of molecular and cellular
cardiology,vol. 51, no. 6, pp. 966–973, 2011.
[202] X. Ou, X. Bin, L. Wang et al., “Myricetin inhibits Kv1.5 chan-
nels in HEK293 cells,”Molecular Medicine Reports, vol. 13,
no. 2, pp. 1725–1731, 2016.
[203] L. Yang, J. H. Ma, P. H. Zhang, A. R. Zou, and D. N. Tu,
“Quercetin activates human Kv1.5 channels by a residue
I502 in the S6 segment,”Clinical and experimental pharma-
cology & physiology, vol. 36, no. 2, pp. 154–161, 2009.
[204] K. H. Chen, H. Liu, H. Y. Sun et al., “The natural flavone aca-
cetin blocks small conductance Ca
2+
-activated K
+
channels
stably expressed in HEK 293 cells,”Frontiers in Pharmacol-
ogy, vol. 8, p. 716, 2017.
[205] K. Singh, A. M. Zaw, R. Sekar et al., “lycyrrhizic acid reduces
heart rate and blood pressure by a dual mechanism,”Mole-
cules, vol. 21, no. 10, p. 1291, 2016.
[206] R. Harikrishnan, G. Devi, H. Van Doan et al., “Study on anti-
oxidant potential, immunological response, and inflamma-
tory cytokines induction of glycyrrhizic acid (GA) in silver
carp against vibriosis,”Fish & Shellfish Immunology,
vol. 119, pp. 193–208, 2021.
[207] K. Chen, R. Yang, F. Q. Shen, and H. L. Zhu, “Advances in
pharmacological activities and mechanisms of glycyrrhizic
acid,”Current Medicinal Chemistry, vol. 27, no. 36,
pp. 6219–6243, 2020.
[208] X. Deng, C. Shen, and Q. Meng, “Screening of herbal compo-
nents for attenuating amiodarone-induced hepatotoxicity on
gel-entrapped rat hepatocytes,”Drug and Chemical Toxicol-
ogy, vol. 37, no. 1, pp. 100–106, 2014.
[209] A. Perino, A. Ghigo, E. Ferrero et al., “Integrating cardiac
PIP
3
and cAMP signaling through a PKA anchoring function
of p110γ,”Molecular Cell, vol. 42, no. 1, pp. 84–95, 2011.
[210] R. Othong, S. Trakulsrichai, and W. Wananukul, “Diospyros
rhodocalyx (Tako-Na), a Thai folk medicine, associated with
hypokalemia and generalized muscle weakness: a case series,”
Clinical Toxicology, vol. 55, no. 9, pp. 986–990, 2017.
[211] T. G. Tolstikova, M. V. Khvostov, A. O. Bryzgalov, I. F.
Belenichev, and S. V. Pavlov, “Glycidipine, a promising
hypotensive and cardioprotective agent,”Bulletin of Exper-
imental Biology and Medicine, vol. 151, no. 5, pp. 597–600,
2011.
[212] A. M. Talman, J. Clain, R. Duval, R. Ménard, and F. Ariey,
“Artemisinin bioactivity and resistance in malaria parasites,”
Trends in Parasitology, vol. 35, no. 12, pp. 953–963, 2019.
[213] S. Zhu, Q. Yu, C. Huo et al., “Ferroptosis: a novel mecha-
nism of artemisinin and its derivatives in cancer therapy,”
Current medicinal chemistry, vol. 28, no. 2, pp. 329–345,
2021.
[214] Y. X. Yan, M. J. Shao, Q. Qi et al., “Artemisinin analogue
SM934 ameliorates DSS-induced mouse ulcerative colitis via
suppressing neutrophils and macrophages,”Acta pharmaco-
logica Sinica, vol. 39, no. 10, pp. 1633–1644, 2018.
[215] S. Rakedzon, A. Neuberger, A. J. Domb, N. Petersiel, and
E. Schwartz, “From hydroxychloroquine to ivermectin: what
are the anti-viral properties of anti-parasitic drugs to combat
22 Cardiovascular Therapeutics
SARS-CoV-2?,”Journal of Travel Medicine, vol. 28, no. 2,
2021.
[216] S. Goswami, R. S. Bhakuni, A. Chinniah, A. Pal, S. K. Kar, and
P. K. Das, “Anti-Helicobacter pylori potential of artemisinin
and its derivatives,”Antimicrobial Agents and Chemotherapy,
vol. 56, no. 9, pp. 4594–4607, 2012.
[217] D. Dolivo, P. Weathers, and T. Dominko, “Artemisinin and
artemisinin derivatives as anti-fibrotic therapeutics,”Acta
Pharmaceutica Sinica B, vol. 11, no. 2, pp. 322–339, 2021.
[218] B. F. Yang, Y. R. Li, C. Q. Xu et al., “Mechanisms of artemisi-
nin antiarrhythmic action,”Chinese Journal of Pharmacology
and Toxicology, vol. 3, pp. 12–18, 1999.
[219] B. F. Yang, C. Q. Xu, Y. R. Li et al., “Inhibition of artemisinin
on cloned inward recirculating potassium channels,”Chinese
Journal of Pharmacology and Toxicology, vol. 4, pp. 245–248,
1999.
[220] Y. Wang and F. Pei, “Artemisinin-regulated Cav 1.2/CaM/
CaMK II pathway in the treatment of atrial fibrillation model
rats,”Journal of Traditional Chinese Medicine, vol. 35, no. 10,
pp. 2182–2189, 2020.
[221] S. Poelzing and D. S. Rosenbaum, “Altered connexin43
expression produces arrhythmia substrate in heart failure,”
Heart and Circulatory Physiology, vol. 287, no. 4,
pp. H1762–H1770, 2004.
[222] Y. Gu, G. Wu, X. Wang, Y. Wang, and C. Huang, “Artemisi-
nin prevents electric remodeling following myocardial infarc-
tion possibly by upregulating the expression of connexin 43,”
Molecular Medicine Reports, vol. 10, no. 4, pp. 1851–1856,
2014.
[223] L. Yin and C. C. Huang, “Research progress on the role of
hyperpolarization-activated cyclic nucleotide gated cation
channels in cardiovascular system,”Guangxi Med, vol. 43,
no. 11, pp. 1371–1375, 2021.
[224] Y. F. Shi, S. J. Chen, Q. H. Wang et al., “Effects of artemisinin
on hyperpolarization-activated cyclic nucleotide gated chan-
nel and pacer current in rabbit sinus atrial node with heart
failure,”Chinese Journal of Heart Failure and Cardiomyopa-
thy,vol. 4, pp. 215–219, 2019.
[225] Q. H. Wang, N. Kuang, W. Y. Hu, D. Yin, Y. Y. Wei, and T. J.
Hu, “The effect ofPanax notoginsengsaponins on oxidative
stress induced by PCV2 infection in immune
cells:in vitroandin vivostudies,”Journal of Veterinary Science,
vol. 21, no. 4, article e61, 2020.
[226] Q. Wu, C. He, X. Wang et al., “Sustainable antibacterial sur-
gical suture using a facile scalable silk-fibroin-based berberine
loading system,”ACS biomaterials science & engineering,
vol. 7, no. 6, pp. 2845–2857, 2021.
[227] J. Sun and R. Mac Kinnon, “Structural basis of human
KCNQ1 modulation and gating,”Cell, vol. 180, no. 2,
pp. 340–347.e9, 2020.
[228] X. Li, S. Sun, D. Chen et al., “Puerarin attenuates the
endothelial-mesenchymal transition induced by oxidative
stress in human coronary artery endothelial cells through
PI3K/AKT pathway,”European Journal of Pharmacology,
vol. 886, article 173472, 2020.
[229] R. Li, Z. L. Song, X. K. Zhang et al., “Progress in research on
pharmacological action and clinical application of Pueraria
root [J/OL],”Journal of Hainan Medical College, vol. 23,
no. 10, pp. 1–16, 2021.
23Cardiovascular Therapeutics