You are on page 1of 15

Molecular Biotechnology (2022) 64:758–772

https://doi.org/10.1007/s12033-022-00449-5

REVIEW

Research Progress of LncRNAs in Atrial Fibrillation


Wenhui Wang1 · Bei Tian2 · Zhongping Ning2 · Xinming Li3 

Received: 13 September 2021 / Accepted: 2 January 2022 / Published online: 2 February 2022
© The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature 2022

Abstract
Atrial fibrillation (AF) is one of the most common arrhythmias in adults, with high morbidity and increased mortality risk. In
recent years, the clinical diagnosis, treatment, and mechanistic research of AF have increased exponentially, and regulation
based on the potential molecular mechanism of AF is a research hotspot. Long noncoding RNAs (LncRNAs), usually refer to
noncoding RNA transcripts greater than 200 nucleotides in length, have been shown to play a role in cardiovascular diseases
such as coronary artery disease, heart failure, and myocardial fibrosis through various regulatory methods. An increasing
number of researchers have begun to pay attention to the identification and function of LncRNAs in AF. This article reviews
changes in the expression of related LncRNAs detected in AF and describes the LncRNAs that play a regulatory role in AF-
related processes, to explore the potential of LncRNAs as new biomarkers and therapeutic targets in AF.

Keywords  Atrial fibrillation · Long noncoding RNA · Differential expression · Biomarker · Therapeutic target

Introduction stroke, dementia, and congestive heart failure (CHF), lead-


ing to serious social and economic burdens [4, 5].
Atrial fibrillation (AF) is one of the most common persistent The current management of AF focuses on symptom
arrhythmias in adults. Among the Framingham Heart Study reduction and complication prevention [6]. Heart rate con-
population, 37% of people have a lifetime risk of develop- trol and rhythm control are used to improve symptoms, and
ing AF for age 55, and men have a higher risk of AF than anticoagulation is used to reduce the risk of stroke. However,
women [1]. The mortality risk in patients with AF is 1.5–1.9 the above treatments are often not sufficiently efficacious and
times that of those without AF [2], and AF can cause sig- are accompanied by potential adverse reactions [7], failing
nificant morbidity and mortality in patients [3]. The global to meet the therapeutic needs of AF patients.
prevalence of AF was 37.574 billion cases (0.51% of the Noncoding RNA (ncRNA) has become a hot topic in car-
global population) by 2017, has increased by 33% in the past diovascular system diseases in recent years. It participates in
20 years and will continue to increase in the next 30 years. the pathophysiological processes of myocardial infarction,
The global burden of AF in 2050 is expected to increase by atherosclerosis, thrombosis, fibrosis, and inflammation and
more than 60% [4]. The prevalence and incidence of AF is a new risk marker for cardiovascular disease [8]. Long
increase with age, and AF is related to a higher incidence of noncoding RNAs (LncRNAs), a group of ncRNAs with
a length greater than 200 nucleotides, have been found to
regulate a variety of diseases, including cancer, Alzheimer’s
disease, and cardiovascular disease [9]. However, fewer
* Xinming Li studies have been found on LncRNA in AF than in other
xinming06@163.com diseases (such as heart failure and atherosclerosis).
1
We hypothesized that LncRNAs play a role in AF and,
Tongji University School of Medicine, Shanghai 200082,
China
therefore, conducted a comprehensive analysis of the current
2
experimental evidence supporting the involvement of LncR-
Department of Cardiology, Shanghai University of Medicine
& Health Sciences Affiliated Zhoupu Hospital, No. 1500 of
NAs in AF with a focus on clinical significance. Background
Zhouyuan Road, Pudong New District, Shanghai 201318, information related to AF and LncRNAs is first presented,
China and the expression of LncRNAs in AF and experimental
3
Shanghai Pudong New Area Center for Disease Control evidence supporting LncRNAs as regulatory factors in AF
and Prevention, Shanghai 200136, China

13
Vol:.(1234567890)
Molecular Biotechnology (2022) 64:758–772 759

are summarized. The goal of this work was to provide a platelet-derived growth factor (PDGF), and connective tis-
comprehensive overview of the research of lncRNA in AF to sue growth factor (CTGF) are effective profibrotic factors
determine the feasibility of LncRNA as potential biomarkers [5]. Inflammation and noncoding RNAs are also involved
and therapeutic targets in AF. in the process of fibrosis [10]. Although the pathogenesis of
AF is controversial and may vary among individuals, elec-
trophysiological abnormalities and atrial structural changes
Pathophysiology of Atrial Fibrillation (including fibrosis) are the bases for the generation and
maintenance of AF [11].
The electrocardiogram has diagnostic value for AF and
reveals the disappearance of the P wave, which is replaced
by an irregular baseline fibrillation f wave, and demonstrates Biology of LncRNAs
an extremely irregular QRS complex [6]. AF can be divided
into three groups based on duration: paroxysmal AF (conver- LncRNAs, noncoding RNAs larger than 200 nucleotides,
sion to sinus rhythm within 7 days), persistent AF (lasting often regulate a variety of diseases, including cancer, Alz-
more than 7 days and needing drugs or electric shock to heimer's disease, and cardiovascular disease [9]. LncRNAs
convert to sinus rhythm), and permanent AF (unsuccess- can be divided into three types based on their positions in
ful conversion to sinus rhythm or relapse within 24 h after the genome: intergenic LncRNAs (lincRNAs), antisense
conversion). The pathogenesis of AF involves many factors, LncRNAs, and intron region LncRNAs [17]. On the LNCi-
including focal ectopic electrical activity, reentry mecha- pedia website (an authoritative summary website of LncR-
nisms, inflammatory mediators, abnormal C ­ a2+ ion chan- NAs), 127,802 transcripts and 56,946 genes are presented.
nels, autonomic nervous system involvement, etc. [10]. The Due to the complexity of the LncRNA population, the bio-
specific mechanisms involved vary depending on the type logical characteristics and mechanisms of most LncRNAs
of AF. are uncertain. Another question that remains unsolved is
The general hypothesis for the occurrence of AF is that whether many LncRNAs have biological functions [17]. It
a rapid electric trigger forms reentrant propagation in the will take many years to elucidate these questions, especially
fragile atrial matrix [11]. Haissaguerre et al. [12] first dis- because many LncRNAs have multiple forms of alternative
covered focal ectopic discharges in muscle sleeve cells along splicing. In an effort to accelerate the progress, researchers
the pulmonary vein (PV) of patients with paroxysmal AF; are using bioinformatics strategies, such as high-throughput
the ablation of these ectopic lesions reduced the burden of sequencing, screening methods, RNA interference (such as
AF, demonstrating their role in the occurrence of AF. After siRNA/shRNA), and target gene prediction, to study the
AF is triggered, a relatively fragile atrial matrix environ- function and mechanism of candidate LncRNAs [18, 19].
ment is formed through electrical remodeling and structural The sequence of LncRNAs is less conserved in mam-
remodeling of the atrium, and the stable reentry mechanism malian LncRNAs, which is similar to that of the intron
promotes the persistence of AF [11]. For patients with long- region of mRNA, but it shows strong conservation in func-
term persistent AF or permanent AF, atrial remodeling is tion [20, 21]. The mechanism of LncRNAs is different from
more extensive and serious, and the presence or absence of that of microRNAs (miRNAs), which directly bind to target
an electric trigger is no longer a decisive factor [11, 13]. A mRNAs to reduce their translation and/or stability [22]. The
brief summary of the pathophysiology of AF is shown in molecular mechanisms of LncRNA are mainly described as
Fig. 1. (i) a bait for localizing transcription factors; (ii) a regulatory
The prominent feature in AF-related electrical remode- signal for transcription; (iii) a scaffold for bridging different
ling is the shortening of the atrial effective refractory period proteins; (iv) a sponge for microRNA; (v) a direction for the
(AERP), which is mainly due to the shortening of the action protein to reach its target; and (vi) possibly possessing short
potential duration (APD) [14], caused by the enhancement of open reading frames (ORFs) that can be translated, allowing
the inward ­K+ current (IK1) or the acetylcholine-dependent it to increase the function of protein [9].
­K+ current (IKACh) or the decrease in the inward L-type ­Ca2+
current (ICaL) [15]. The development of cardiac structural
remodeling, characterized by atrial enlargement and fibrosis, Expression of LncRNA in AF Patients
is slower than electrical remodeling [15]. Atrial enlargement
helps maintain the complex mechanism of reentrant arrhyth- There are two types of evidence for the pathophysiological
mia, and atrial fibrosis contributes to the atrial matrix of role of LncRNAs in AF. First, abnormal LncRNA expression
AF [16]. However, the mechanism of atrial fibrosis is not is often observed in the heart tissue and blood of AF patients
fully understood. Multiple studies have shown that angio- and animal models. The changes in heart tissue indicate a
tensin-II (Ang-II), transforming growth factor β1 (TGF-β1), potential role in AF-related cardiac remodeling, while the

13

760 Molecular Biotechnology (2022) 64:758–772

changes in blood suggest that LncRNAs may have diagnostic Xu et al. [30] also found some DELncRNAs in smaller AF
and prognostic value. Second, experiments have confirmed populations.
that LncRNAs have a direct causal relationship with AF by Su et al. [28] found that LncRNA RP11-519G16.3 was
manipulating LncRNA expression, suggesting the therapeu- upregulated in 31 paroxysmal AF patients compared to
tic potential of LncRNAs in AF. controls, and AK000451 was downregulated. This sug-
gests that these two LncRNAs may contribute to predict-
ing the occurrence of paroxysmal AF. Yu et al. [29] found
The Expression of LncRNAs in the Blood of AF 49 upregulated and 45 downregulated LncRNAs in 6 per-
Patients sistent AF patients and 6 controls. The study shows that
higher levels of TCONS_00024161, TCONS_00194688,
Researchers [23–30] collected blood samples from patients and TCONS_00076385 may be useful to predict persistent
with AF and control groups with sinus rhythm and per- AF. A bioinformatics analysis was conducted on the dif-
formed RNA sequencing, microarray, and/or polymerase ferentially expressed mRNAs and LncRNAs and identified
chain reaction (PCR) methods to screen out the differen- enriched signaling pathways, such as the TNF signaling
tially expressed LncRNAs (DElncRNAs) in AF patients. The pathway, NF-κB signaling pathway, Toll-like receptor path-
results showed an abundant expression profile change. Some way, and NOD-like receptor pathway. These pathways are
studies further explored the clinical and practical values of related to apoptosis, immunity, inflammation, and oxidative
DELncRNAs for AF patients through clinical follow-up. The stress, suggesting that these signal transduction pathways
experimental designs and results of these studies are shown may play an important role in the initiation and development
in Table 1. of AF [29].
Qian et al. [31] recommended LncRNA GAS5 as a new In prognostic studies of AF patients, Zeng et al. [24]
biomarker in atrial tissue for AF by constructing an AF- followed patients for 2–4 years and found that AF patients
related LncRNA-mRNA network (AFLMN) via microar- without ischemic stroke had lower serum LncRNA ANRIL
ray analysis, which shows powerful diagnostic ability for levels. The sensitivity and specificity of LncRNA ANRIL
AF. Functional module analysis shows that GAS5 is closely for recognizing ischemic stroke with AF were 76.6% and
related to 52 mRNAs and is enriched in many AF-related 81.4%, respectively.
pathways involved in inflammation, ion channels, and meta- Overall, these results show that circulating LncRNA
bolic regulation. Another study [32] also confirmed the sig- changes in AF patients, but inconsistent results indicate that
nificant difference in the expression of GAS5 in the atrial LncRNAs are not currently strong biomarkers for AF. Part of
tissue of AF patients and emphasized its inhibitory effect the problem may be the heterogeneity of the AF population,
on proliferation. Shi et al. [23] tested the level of LncRNA and more specific subgroups (such as isolated AF patients,
GAS5 in the blood of AF patients and found that its expres- AF patients with congestive HF, or AF patients only with
sion was significantly downregulated. This change occurs hypertension) may need to be studied. Another problem is
before the left atrial volume enlarges and is closely related that the blood samples in these studies were different, and
to the progression of AF, indicating the potential role of the presence or absence blood cells will cause great differ-
GAS5 as an early biomarker for AF. In the group of patients ences in results. In the future, LncRNAs could be identified
with persistent AF without a history of hypertension, blood as reliable biomarkers in AF. These results can be useful for
GAS5 was further downregulated, indicating that GAS5 can risk stratification, individualized treatment based on patho-
be used for the prognosis of AF progression. Of the 70 AF physiology, evaluation of AF progression, and assessment
patients who underwent radiofrequency catheter ablation of response to treatment.
(RFCA), 22 (31.4%) experienced recurrence at the 1-year
follow-up. K-M analysis (log-rank test, P = 0.031) and mul- The Expression of LncRNAs in Heart Tissue of AF
tivariate Cox analysis (HR = 0.127, 95% CI 0.026–0.616; Patients
P = 0.01) showed that GAS5 may predict the recurrence of
atrial arrhythmia after RFCA. Currently, RNA sequencing and microarray analysis, as out-
Wang et al. [27] studied the blood LINC00472 levels in standing disease mining technologies, have been applied to
125 AF patients and 168 control patients and found that the identify DELncRNAs in AF patients and healthy people.
expression was decreased in AF patients, while the DNA Researchers [32–43] collected heart tissue samples from
methylation level of LINC00472 was increased. Yao et al. patients with AF and control groups with sinus rhythm and
[25] evaluated the differential expression of LncRNA MIAT performed the same methods as above to screen out the
in the white blood cells of 35 AF patients and 30 healthy DElncRNAs in heart tissue of AF patients. The heart tis-
controls and found that the expression of MIAT was sig- sue samples included atrial tissue, epicardial adipose tissue,
nificantly upregulated in AF patients. Ruan et al. [26] and and pericardial fluid of AF patients. As expected, the initial

13
Table 1  The expression of LncRNAs in the blood of AF patients
Studies Patients Samples Methods Results

Shi et al. [23] (screening) AF (n = 20) and control (n = 20); Plasma qRT-PCR (screening): Reduced level of blood LncRNA GAS5
(validation) AF (n = 85) and control (n = 43); was observed in patients with AF. Other three candi-
1-year follow-up of patients with AF underwent radio dates (LncRNA NEAT1, UCA1 and TUG1) had not
frequency catheter ablation (RFCA) (n = 70) been observed significant difference
(validation): Reduced level of circulating LncRNA GAS5
was observed in patients with AF
1-year follow-up of patients with AF underwent RFCA:
K-M analysis showed that the low GAS5 group had a
higher risk of recurrence after RFCA. Multivariate anal-
ysis showed that GAS5 and left ventricular end diastolic
Molecular Biotechnology (2022) 64:758–772

diameter were independent predictors of recurrence


Zeng and Jin [24] 2–4 years follow-up of patients with AF: AF with Serum qRT-PCR Reduced level of blood LncRNA ANRIL was observed in
ischemic stroke (n = 132) and AF without ischemic AF patients without ischemic stroke compared with the
stroke (n = 254) patients that have AF and ischemic stroke
Yao et al. [25] AF (n = 35) and control (n = 30) Peripheral leukocyte qRT-PCR LncRNA MIAT was upregulated in peripheral leukocytes
of AF patients
Ruan et al. [26] AF (n = 20) and control (n = 20) Peripheral monocyte Microarray analysis, Upregulated: HNRNPU-AS1*, LINC00926,
qRT-PCR LINC00861*, AC005786.7, (AC005786.7),
XLOC_099061
Downregulated: GS1-115G20.1, LINC00877,
ENSG00000239794, LINC00476, CTD-2616J11.3,
CTD-2616J11.2*, RP11-443B7.3*, CTD-2006K23.1,
(CTD-2616J11.14), LINC00936, RP11-810P12.7,
XLOC_060065, XLOC_109634
Wang et al. [27] AF (n = 125) and control (n = 168) Plasma qRT-PCR The expression level of LINC00472 was decreased in
patients with AF, while the DNA methylation level of
LINC00472 was increased
Su et al. [28] Paroxysmal AF (n = 3) and control (n = 3); Peripheral leukocyte Microarray analysis, qRT-PCR Upregulated: HBBP1, RP11-119D9.1, RP11-519G16.3*,
(validation) paroxysmal AF (n = 31) and control (n = 31) LOC100130275, XLOC_006934, (DQ583499),
AC139099.4, XLOC_005160, RPL29P30, RP11-
40A13.1, AC073135.3, RP5-981L23.1, LOC283761,
RP11-64I17.1, XLOC_002560, RP11-328K4.1,
BC040700, AC131097.3, RP11-16,217.1
Downregulated: (RP11-605F22.2), XLOC_007697,
RP11-1060J15.4, PI4KAP2, AK000451*, CTD-
2574D22.4, TTTY6, RP1-251I12.1, XLOC_005775,
XLOC_010596, AY927499, chr12:53,675,775–
53,689,275, BC160930, RNF157-AS1, XLOC_007777,
LOC375196, PRSS21
Validation: RP11-519G16.3 and AK000451 may be help-
ful in predicting paroxysmal AF

13
761

Table 1  (continued)
762

Studies Patients Samples Methods Results

13
Yu et al. [29] Persistent AF (n = 6) and control (n = 6) Peripheral lymphocyte RNA sequencing, qRT-PCR Upregulated: TCONS_00039400, TCONS_00008957,
TCONS_00024161*, TCONS_00194688*,
TCONS_00024160, TCONS_00045457,
TCONS_00164108, TCONS_00064672,
TCONS_00076385*, TCONS_00128364 and etc
Downregulated: TCONS_00202959, TCONS_00065233,
TCONS_00049962, TCONS_00134390,
TCONS_00095749, TCONS_00147447,
TCONS_00028780, TCONS_00065235,
TCONS_00010002, TCONS_00132941 and etc
Xu et al. [30] AF (n = 3) and control (n = 3); Whole blood Microarray analysis, Upregulated: NONHSAT098586, NONHSAG007503*,
(validation) AF (n = 20) and control (n = 10) qRT-PCR NONHSAT076398, NONHSAT053927,
NONHSAT098582, NONHSAT017667,
NONHSAT090718, NONHSAT098591,
NONHSAT076870, NONHSAT076401,
NONHSAT055019
Downregulated: NONHSAT040387*, NONHSAT040421,
TCONS_l2_00030433, NONHSAT039492,
NONHSAG053956, NONHSAT136136,
XR_245045.1, NONHSAT141981, NONHSAT142052,
NONHSAT040390, ENST00000460164,
ENST00000456563, NONHSAT040540,
NONHSAT136135

AF atrial fibrillation, qRT-PCR quantitative real-time polymerase chain reaction


*LncRNA indicates dysregulated LncRNA that has statistically significant changes on subsequent qRT-PCR measurement. Bracketed LncRNA antisense
Molecular Biotechnology (2022) 64:758–772
Molecular Biotechnology (2022) 64:758–772 763

research data indicate that the number of DELncRNAs in the the electrical and structural remodeling of AF [46]. Further
heart tissue of AF patients is significantly greater than that in exploration of gene expression and regulatory mechanisms
blood. The differential expression of LncRNAs in the heart in the heart will help reveal new treatments for AF. In the
tissues of AF patients (see Table 2 for details) suggests that next section, we summarize the LncRNAs that are kown to
the DELncRNAs may be related to AF. play regulatory roles in AF.
Many researchers have conducted differential expres-
sion studies in the atria, epicardial adipose tissue, and
pericardial fluid of AF patients and have reported exciting Mechanisms of LncRNAs in AF
results. LncRNA GAS5 was also found to be downregu-
lated in the atrial tissue of AF patients [32], making it an To date, LncRNAs have been used as new biomarkers in
LncRNA that has been described in both the circulation and many areas, such as tumors, neurological diseases, and
heart tissues of AF patients. At the same time, NEAT1 was cardiovascular diseases (CVDs) [47]. Various studies have
found to be upregulated in the right atrial appendage tis- shown that the dysregulation of LncRNA expression is rel-
sue of AF patients [35], but no significant difference was evant to AF. Some LncRNAs and mRNAs share the same
observed in blood samples [23], suggesting that there is dif- miRNA. Therefore, LncRNAs act as "sponges" of miRNAs
ferential expression of LncRNA in tissues and blood in AF and can regulate mRNA transcription [48], which suggests
patients. The details of different studies warrant attention. that LncRNAs may play a role in signal transduction in the
LINC00636 was found to be downregulated in the human pathogenesis of AF. The AF-associated LncRNAs that act
pericardial fluid of AF patients [33], but no studies have as "sponges" of miRNAs are shown in Fig. 2.
identified it in other tissues of AF patients. The common experimental design to explore the mecha-
Ke et al. [38] conducted a sequencing screen on the paired nism of LncRNAs in AF is to select a target LncRNA that
left and right atrial appendages of 5 AF patients and 5 non- was differentially expressed in the AF patient or AF animal
AF patients and analyzed several related LncRNAs. NPPA- model and then to observe the effect by inhibiting or upregu-
AS1 may participate in the pathogenesis of AF by regulating lating the expression of the LncRNA in an animal model
muscle contraction. RP11-99E15.2 interacts with the protein (in vivo) or in a cell model (in vitro). Electrical remodeling
ITGB3 and participates in ECM-receptor interaction and the and structural remodeling are commonly observed. Even the
intracellular activation of TGF-β signal transduction. The relatively rare LncRNAs that can affect autonomic nerve
combination of RP3-523K23.2 and the protein HSF2 may remodeling are ultimately involved in atrial electrical remod-
enhance the transcriptional activity of HSF2, thereby reshap- eling by affecting AERP [49].
ing ion channels by regulating the transcription of HSF2 As mentioned above, the increase in IK1 and the decrease
target genes. in ICaL are the two most significant changes in ionic channels
Tissue-specific gene expression programs are carefully in AF-associated electrical remodeling. Table 3 shows the
controlled during heart development [44]. Yeh et al. [45] LncRNAs related to AF electrical remodeling. Atrial fibro-
reported that there are 391 differentially expressed genes sis is a sign of AF structural remodeling. Table 4 shows the
between left atrium (LA)-PV and left atrial appendage LncRNAs related to the atrial structural remodeling in AF.
(LAA) in patients with persistent AF, which are related to One study [50] found that TCONS_00075467 was down-
arrhythmia, cell death, inflammation and hypertrophy. Chen regulated in the AF rabbit model and could sponge miR-
et al. [42] further found 94 DELncRNAs between LA-PV 328 as an endogenous competitive RNA (ceRNA). Over-
and LAA in AF patients by microarray analysis. Among expression of miR-328 increased susceptibility to AF [51]
these LncRNAs, AK055347 was selected for further verifi- by reducing the voltage-dependent L-type calcium channel
cation in H9C2 cardiomyocytes. LncRNAs may deregulate current (ICaL), reducing the expression of the L-type cal-
mitochondrial energy production by regulating Cyp450, ATP cium channel α subunit and β subunit, and shortening APD.
synthase, and MSS51, thereby promoting the pathogenesis This shows that downregulation of TCONS_00075467 has
of AF. Knockdown of AK055347 significantly inhibited the arrhythmogenic effects.
viability of H9C2 cardiomyocytes. Therefore, AK055347 The LncRNA KCNQ1OT1 was found to be upregulated
upregulates and regulates mitochondrial energy production in the AF mouse model [52]. The mechanism of KCN-
in cardiomyocytes in AF patients. It is well known that the Q1OT1 in AF is to regulate the expression of CACNA1C
LA-PV area is an important area where electrophysiological by sponging miR-384 [52]. Silencing KCNQ1OT1 can pro-
abnormalities of AF originate. Electrical triggering remains long AERP, shorten interatrial conduction time (IACT),
an important initiating factor for patients who have not yet and improve atrial electrical remodeling, which produces a
reached the permanent AF stage [11, 13]. This means that therapeutic effect on AF.
intervention of the origin of electrophysiological abnormali- The expression of TCONS-00106987 was upreg-
ties can greatly reduce the electrical load of AF and improve ulated in the AF group [53]. Overexpression of

13

Table 2  The expression of LncRNAs in heart tissue of AF patients


764

Studies Patients Samples Methods Results

13
Liu et al. [33] AF (n = 12) and control (n = 12) Exosomes in human pericardial fluid qRT-PCR Compared with control group, reduced level of
LINC00636 was observed in exosomes of AF
patients’ pericardial fluid
Sun et al. [34] Persistent AF (n = 3), paroxysmal AF (n = 3) and LAA RNA sequencing, qRT-PCR Compared with control group, reduced level of
control (n = 3) RP11-442H21.2 was observed in LAA of parox-
ysmal AF patients, while reduced level of CTA-
134P22.2 was observed in persistent AF patients
Dai et al. [35] AF (n = 15) and control (n = 15) RAA​ qRT-PCR LncRNA NEAT1 was upregulated in RAA of AF
patients
Lu et al. [24] AF (n = 40) and control (n = 30) RAA​ qRT-PCR LncRNA GAS5 was downregulated in RAA of AF
patients
Wu et al. [36] Patients with valvular heart disease and AF RAA​ RNA sequencing, qRT-PCR Upregulated: LOC105370977, LOC105371928,
(n = 7) and patients with valvular heart disease LOC105376344, BANCR, SEC1P,
but no AF (n = 7); LOC105369818, LOC440568, LOC105377983,
(validation): Patients with valvular heart disease VWFP1, LINC01479, etc
and AF (n = 35) and patients with valvular heart Downregulated: IMMTP1, LOC105374598,
disease but no AF (n = 35) LGALS17A, KRT88P, LOC105373923,
LOC105375472, LOC105372145,
LOC101928989, LOC105377227,
LOC105372685, etc
(validation): VDAC2P2* was found to have signifi-
cantly difference among three selected LncRNAs
Zhao et al. [37] Non-valvular persistent AF (n = 6) and control EAT Microarray analysis, qRT-PCR Upregulated: TPRG1, PDLIM1*, AC015802.6,
(n = 6) ANKRD12, SUB1, NOS3, AHCTF1, LAMC1,
FAM208B, MRPL43, TTC3, MSANTD2,
MAP3K7CL, TBRG1, ENST00000489394*,
ENST00000557365*, etc
Downregulated: CTSB, FXYD6, LGALS3*, PI16,
GPX3, ICAM2, ANAPC5, WNT11, RASSF4,
GPAA1, LOXL3, DNM1, FCGR2C*, PRMT7,
NUBP1, ENST00000491449*, etc
Ke et al. [38] AF (n = 5) and control (n = 5) LAA, RAA​ RNA sequencing The biological changes of AF between LAA and
RAA were similar
Wu et al. [39] Patients with rheumatic valvular heart disease and LAA, RAA​ Microarray analysis LncRNA-n336928 was the most significantly
AF (n = 7) and patients with rheumatic valvular upregulated LncRNA in AF patients
heart disease but no AF (n = 10)
Wang et al. [40] AF (n = 34) and control (n = 20) Atrial tissue qRT-PCR LncRNA NRON was downregulated in the atria of
AF patients
Molecular Biotechnology (2022) 64:758–772
Table 2  (continued)
Studies Patients Samples Methods Results

Mei et al. [41] Patients with rheumatic valvular heart disease and RAA​ Microarray analysis, qRT-PCR Upregulated: uc010vaf.1, DQ596229,
AF (n = 3) and patients with rheumatic valvular TCONS_00023347, DQ589437, SNORD115-
heart disease but no AF (n = 3); 22*, SNORD115-38, SNORD115-32,
(validation): patients with rheumatic valvular SNORD115-42, CR608741, SNORD115-6, etc
heart disease and AF (n = 15) and patients with Downregulated: uc001eiy.2, uc001ejh.1,
rheumatic valvular heart disease but no AF DQ590126, DQ579288, BC041938*,
(n = 15) DQ576791, DQ576039, uc010yty.1, DQ595787,
TCONS_00005387, etc
Chen et al. [42] Persistent AF (n = 16) Pulmonary veins and the surround- Microarray analysis, qRT-PCR The top 10 LncRNAs with most significant differ-
ing left atrium area (LA-PV), LAA ential expression between LA-PV and LAA in AF
Molecular Biotechnology (2022) 64:758–772

patients: AK055347*, AK310040, AK026494,


BC010527, AK027294, AB007979, AK091573,
UC003qev.1, AK125186, U9981
Ruan et al. [43] Patients with rheumatic valvular heart disease and LAA Microarray analysis, qRT-PCR Upregulated: ENST00000575612*, uc001eqh.1*,
AF (n = 3) and patients with rheumatic valvular BC064139*, ENST00000425309*,(E
heart disease but no AF (n = 3) NST00000500112)
Downregulated: TCONS_00006371*, Z74666*,
X85157*,(X72487), NR_033661*

AF atrial fibrillation, qRT-PCR quantitative real-time polymerase chain reaction, LAA left atrial appendage, RAA​right atrial appendage, EAT epicardial adipose tissue
*LncRNA indicates dysregulated LncRNA that has statistically significant changes on subsequent qRT-PCR measurement. Bracketed LncRNA antisense

13
765

Table 3  LncRNA associated with electrical remodeling of AF


766

LncRNA Model Pathway Results

13
HOTAIR Chronic AF patients; miR-613/ Cx43 HOTAIR was downregulated in chronic AF patients,
HL-1 cardiomyocytes and the level of connexin 43 (Cx43) mRNA and
Cx43 protein were also downregulated
In vitro, HOTAIR positively regulated Cx43 by spong-
ing miR-613, and the decrease of Cx43 was related to
atrial electrical remodeling [55]
KCNQ1OT1 AF mouse model; miR-384/ CACNA1C In AF mouse model, four LncRNAs were signifi-
Ang-II-induced mouse cantly upregulated: KCNQ1OT1, LOC102723321,
KCNQ1-AS1, KCNJ2-AS1
Among them, only KCNQ1OT1 was upregulated in
both AF mouse model and Ang-II- induced mouse
heart [52]
In vivo, silencing KCNQ1OT1 prolonged AERP,
shorten IACT, and reduced the AF induction rate and
AF duration that was caused by Ang-II. Inhibiting
KCNQ1OT1 could improve atrial electrical remod-
eling
TCONS-00106987 AF rabbit model of atrial rapid pacing miR-26/ KCNJ2 TCONS-00106987 was upregulated in the AF rab-
bit model Overexpression of TCONS-00106987
significantly shortened AERP and increased the AF
induction rate, while inhibition leaded to the opposite
effect
In vivo, TCONS-00106987 induced the transcription
of KCNJ2 by sponging miR-26, thereby increas-
ing the inward rectifier K channel current ­(IK1) and
promoting electrical remodeling [53]
MIAT AF patients; miR-133a-3p MIAT was upregulated in the blood leukocytes of AF
AF rat model of atrial rapid pacing patients
In vivo, the expression of MIAT increased significantly
after AF induction, and showed a gradual downward
trend over time
MIAT could sponge miR-133a-3p to play a regulatory
role Inhibition of MIAT increased AERP, shortened
the duration of AF, and reduced AF-induced cardio-
myocyte apoptosis and atrial fibrosis [25]
LncRNA TCONS_00202959 AF rat model of Ach-CaCl2 Unclear TCONS_00202959 was downregulated in the AF rat
model
Overexpression of TCONS_00202959 prolonged
AERP, reduced the AF induction rate, and improved
electrical remodeling in rats [56]
Molecular Biotechnology (2022) 64:758–772
Molecular Biotechnology (2022) 64:758–772 767

TCONS-00106987 significantly shortens AERP and

AF atrial fibrillation, AERP atrial effective refractory period, IACT​ interatrial conduction time, APD action potential duration, ceRNA competing endogenous RNAs, Ang-II angiotensin-II, ICaL
were screened out to be possibly related to autonomic
1220 DELncRNAs were identified by RNA sequencing

Silencing TCONS_00075467 shortened AERP in vivo,

In vitro and in vivo, TCONS_00075467 could regulate


the expression of CACNA1C by sponging miR-328
in AF rabbit model, and the expression of LncRNA

In vivo, knockdown of TCONS_00032546 shortened


increases the AF induction rate, while inhibition leads

prolonged AERP. Both of them participate in atrial


were identified by sequencing in AF canine model
TCONS_00032546, TCONS_00026102 AF canine model of atrial rapid pacing The former may activate the MAPK pathway through 410 upregulated and 166 downregulated LncRNAs

Through target gene prediction, the two LncRNAs


to the opposite effect. An in vivo study has shown that

AERP, and knockdown of TCONS_00026102


TCONS_00075467 was downregulated [50]
TCONS-00106987 forms endogenous competition by

electrical remodeling in different ways [49]


sponging miR-26, and previous studies have confirmed

and regulate atrial electrical remodeling


that KCNJ2 (a gene encoding the Kir2.1α subunit of the
and decreased ICaL and APD in vitro
inward rectifier potassium channel) is the target gene of
miR-26 [54]. Upregulated TCONS-00106987 induces
the transcription of KCNJ2 by sponging miR-26, thereby
increasing the inward rectifier K channel current ­(IK1) and

neural remodeling
promoting electrical remodeling [53].
LncRNA HOTAIR was downregulated in chronic AF
patients, and it was confirmed in cardiomyocytes that
HOTAIR can positively regulate Cx43 by sponging miR-
Results

613 [55], indicating that dysregulated HOTAIR contributes


to the pathogenesis of AF. However, further in vivo experi-
ments are essential to establish direct causality.
and the latter may increase SLC25A4 through the

Zhao et  al. [56] found that the DELncRNA


the CCND1-FGF19-FGF4-FGF3 gene cluster,

TCONS_00202959 is related to electrical remodeling in an


AF rat model, which has been observed to be consistent
with circulating lymphocytes in AF patients [29]. Overex-
pression of TCONS_00202959 prolonged the AERP in rats.
Wang et al. [49] screened two LncRNAs related to elec-
trical remodeling in the AF canine model and confirmed
the effects on electrical remodeling by knocking down the
miR-328/ CACNA1C

LncRNAs, which has shown potential for further clinical


NF-κB pathway

transformation.
Yao et al. [25] found that inhibiting MIAT increased
AERP and improved cardiac electrical remodeling, while
Pathway

reducing AF-induced cardiomyocyte apoptosis and atrial


fibrosis, but they did not further investigate the mechanism.
MIAT can bind with miR-133a-3p to inhibit its expression,
AF rabbit model of atrial rapid pacing

but the role of miR-133a in cardiac electrical remodeling is


not yet clear. Some studies have found that miR-133a inhibits
the expression of the IKr channel (a repolarization potassium
current channel) encoded by ether-a-go-go-related genes
(ERGs), leading to pathological prolongation of the QT
interval [57]. Therefore, we predict that MIAT may increase
AF sensitivity by affecting the inward potassium current dur-
ing repolarization. On the other hand, MIAT upregulation
Model

increases atrial fibrosis in AF animals by sponging miR-


133a-3p. Inhibiting it can improve atrial fibrosis and AF
structural remodeling. The signal pathways are intricately
related [58]. A literature review found that miR-133 can
directly downregulate the expression of TGF-β1 [59] and
L-type calcium channel current

can also bind to the 3' untranslated region of CTGF mRNA


to reduce the expression of CTGF, a downstream molecule
of the TGF-β/Smad pathway [60]. Liu et al. [61] constructed
Table 3  (continued)

TCONS_00075467

an LncRNA/mRNA gene coexpression network based on the


RNA sequencing results of 235 left atrial appendage samples
and identified MIAT and LINC00964 as key LncRNAs that
LncRNA

participate in multiple pathways of cardiac remodeling in


AF patients.

13

768 Molecular Biotechnology (2022) 64:758–772

Table 4  LncRNA associated with structural remodeling of AF


LncRNA Model Pathway Results

NEAT1 Persistent AF patients; miR-320/NPAS2 The expression of NEAT1 was upregulated in


Ang-II-induced atrial fibrosis mouse model patients with AF, and was positively correlated
with the level of collagen I and collagen III
In vivo, silencing NEAT1 inhibited atrial fibrosis
caused by Ang-II
NEAT1 positively regulated the expression of
NPAS2 (neuronal PAS domain protein 2) by
sponging miR-320 [35], which is related to
atrial fibrosis
NRON AF patients; NFATc3 NRON was downregulated in atrial tissues of AF
Human atrial fibroblasts treated with Ang-II; patients
Ang-II-induced atrial fibrosis mouse model In vitro, overexpression of NRON inhibited the
proliferation of human atrial fibroblasts
In vivo, overexpression of NRON promoted
NFATc3 phosphorylation in Ang-II-induced
mice and inhibited cardiac fibrosis [40]
NRON Atrial myocytes of mouse NFATc3 /IL-12 In vitro, NRON inhibited activation of M1 mac-
rophages from atrial myocytes, and alleviated
atrial fibrosis [62]
NRON Fibroblasts and atrial myocytes of mouse miR-23a In vitro, NRON inhibited miR-23a, which is
derived from atrial myocytes, stimulated M2
macrophages polarization, and alleviated atrial
fibrosis [63]
LICPAR Human atrial fibroblasts from patients with AF TGF-β/Smad signaling pathway In vitro, Ang-II treatment increased the levels
and SR of cell LIPCAR, Smad2/3 phosphorylation,
and α-smooth muscle actin (α-SMA) Overex-
pression of LIPCAR promoted the levels of
collagen I, collagen II, α-SMA, and Smad2/3
phosphorylation, promoted cell viability, and
atrial fibroblast proliferation, while silencing
LIPCAR leaded to the opposite effect [69]
PVT1 Human atrial muscle tissue from patients with miR-128-3p/SP1/TGF-β1/Smad PVT1 was upregulated in the atrial tissue of AF
AF and SR; patients [65]
Ang-II-induced atrial fibrosis mouse model In vivo, PVT1-knockdown attenuated Ang-II-
induced atrial fibrosis in mice. In mechanism,
PVT1 activated the TGF-β/Smad signaling
pathway by sponging miR-128-3p and promot-
ing Sp1 expression [65]. Inhibition of PVT1 can
reduce atrial fibrosis
MEG3 AF canine model of atrial rapid pacing DLK1-DIO3 locus Through the gene sequencing of the AF dog
(1 week); model, LncRNA MEG3 was found to be dys-
AF canine model of atrial rapid pac- regulated at the DLK1-DIO3 locus [66]
ing + AVB + ventricular pacing (1 week) Previous studies have shown that MEG3 can
inhibit TGF-β genes and alleviate the fibrotic
response [67]
MIAT AF patients and healthy individuals; miR-133a-3p MIAT was upregulated in the blood leukocytes of
AF rat model of atrial rapid pacing AF patients
In vivo, the expression of MIAT increased signifi-
cantly after AF induction and showed a gradual
downward trend over time
MIAT could sponge miR-133a-3p to play a
regulatory role. Inhibition of MIAT increased
AERP, shortened the duration of AF, and
reduced AF-induced cardiomyocyte apoptosis
and atrial fibrosis [25]

AF atrial fibrillation, SR sinus rhythm, Ang-II angiotensin-II, α-SMA α-smooth muscle actin, AVB atrial ventricular block, AERP atrial effective
refractory period

13
Molecular Biotechnology (2022) 64:758–772 769

Fig. 1  The pathophysiology of AF

Fig. 2  A brief summary of AF-associated LncRNAs

Studies have found that the expression of LncRNA 63]. It can inhibit the activation of atrial macrophages
NRON is decreased in the atrial tissues of AF patients and reduce atrial fibrosis through inflammatory path-
[40]. Overexpression of NRON can reduce atrial fibrosis ways [62, 63]. It can also promote the phosphorylation of
in AF mouse models through different pathways [40, 62, NFATc3 (nuclear factor 3 of activated T cells) and inhibit

13

770 Molecular Biotechnology (2022) 64:758–772

the activity of activated T cell factor (NFAT) to reduce Conclusions


atrial fibrosis [40]. Further studies found that NRON is
abnormally expressed in the serum of patients with heart The novel proposal in this review is the use of LncRNAs as
failure [64], suggesting that LncRNAs can also be used AF biomarkers, which updates the field of clinical applica-
as circulating biomarkers and have excellent prospects in tion. Studies have shown that circulating LncRNAs have
the cardiovascular field. diagnostic and prognostic value and can be used as early
LncRNA PVT1 activates the downstream TGF-β/ biomarkers of AF, prognostic prediction of AF, and pre-
Smad pathway to promote atrial fibrosis by regulating diction of the recurrence of atrial arrhythmia after RFCA.
miR-128-3p-SP1 in patients with AF [65]. Inhibition of GAS5 and ANRIL, for example, are described in the article.
PVT1 can alleviate atrial fibrosis; silencing of LncRNA Multiple studies have shown that LncRNAs exert impor-
LICPAR also has a therapeutic effect on AF by affecting tant contributions to the pathogenesis of AF. LncRNAs can
the TGF-β/Smad pathway. Through gene sequencing of be used as new targets to interfere with the occurrence and
the AF dog model, it was found that the DLK1-DIO3 development of AF, new AF treatments can be developed
locus was transcriptionally activated after 1 week of con- based on LncRNAs, while circulating LncRNAs can be used
tinuous arrhythmia, and the LncRNA MEG3 was found as novel diagnostic and prognostic markers. At present, more
to be dysregulated at this locus [66]. MEG3 can inhibit research is needed to clarify the “identity” of LncRNAs in
TGF-β genes and alleviate the fibrotic response [67]. The AF and transform this information into clinical practice.
previous work of this laboratory in the AF canine model
showed that maintaining AF stimulation for 1 week acti- Acknowledgements  The literature search was completed in June
vated fibroblasts, collagen gene expression, and changes 2021. We apologize to colleagues whose work was not cited in this
review due to page limitations. All authors read and approved the final
in ion channels in cardiomyocytes by rapid atrial pacing manuscript.
but has not yet caused fibrosis [68]. Continued stimula-
tion for 3 weeks promoted fibrosis. Although no obvi- Author contributions  All authors contributed to the study conception
ous atrial fibrosis was observed in the 1-week AF animal and design. WW and BT had the idea for the article, WW performed
model, there were disorders of fibrosis-related genes; we the literature search and data analysis, ZN and XL critically revised the
work. All authors read and approved the final manuscript.
can infer that if the gene regulation of the myocardial
fibrosis pathway is carried out in the early stage of AF, it Funding  This research was funded by the Key Specialty of Disci-
may have an early intervention effect on AF. pline Construction Project of Shanghai Health Committee (Grant No.
Although the potential therapeutic value of LncRNAs ZK2019B25), and the Research Project of Science and Technology
has been proven in vivo and in animal studies, their long- Commission of Pudong New Area (Grant No. PKJ2019-Y40).
term efficacy, safety and pharmacokinetics in humans are
still largely unknown. Therefore, more research is needed Declarations 
to develop clinically approved therapies for LncRNAs.
Conflict of interest  The authors declare no conflicts of interest.

Research Limitations and Perspectives


References
One limitation is that research on LncRNAs is still in 1. Staerk, L., et  al. (2018). Lifetime risk of atrial fibrillation
the early stage. Most LncRNAs do not yet have clearly according to optimal, borderline, or elevated levels of risk fac-
defined functions, and even the naming methods of LncR- tors: Cohort study based on longitudinal data from the Framing-
NAs have not been fully unified. With further research, ham Heart Study. BMJ, 361, k1453.
2. Benjamin, E. J., et al. (1998). Impact of atrial fibrillation on
some genes originally classified as LncRNAs may be the risk of death: The Framingham Heart Study. Circulation,
found to have protein coding functions and exit the clas- 98(10), 946–952.
sification. However, the studies described in this article 3. Chung, M. K., et al. (2020). Atrial fibrillation: JACC council
were all published after 2015, cover a short time span perspectives. Journal of the American College of Cardiology,
75(14), 1689–1713.
and exhibit more consistent results. Another limitation is 4. Lippi, G., Sanchis-Gomar, F., & Cervellin, G. (2021). Global
that more DELncRNAs described in this article have not epidemiology of atrial fibrillation: An increasing epidemic and
undergone further experimental validation because of rap- public health challenge. International Journal of Stroke, 16(2),
idly increased data. In this article, the verified LncRNAs 217–221.
5. Li, C. Y., et al. (2021). Atrial fibrosis underlying atrial fibrillation
were annotated, and other results will be updated if there (Review). International Journal of Molecular Medicine, 47(3), 9.
is progress in follow-up research. 6. Michaud, G. F., & Stevenson, W. G. (2021). Atrial fibrillation.
New England Journal of Medicine, 384(4), 353–361.

13
Molecular Biotechnology (2022) 64:758–772 771

7. Packer, D. L., et al. (2019). Effect of catheter ablation vs antiar- 29. Yu, X. J., et al. (2017). Long noncoding RNAs and novel inflam-
rhythmic drug therapy on mortality, stroke, bleeding, and cardiac matory genes determined by RNA sequencing in human lympho-
arrest among patients with atrial fibrillation: The CABANA ran- cytes are up-regulated in permanent atrial fibrillation. Am J Transl
domized clinical trial. JAMA, 321(13), 1261–1274. Res, 9(5), 2314–2326.
8. Thomas, M. R., & Lip, G. Y. (2017). Novel risk markers and risk 30. Xu, Y., et al. (2016). Identification of long non-coding RNAs as
assessments for cardiovascular disease. Circulation Research, novel biomarker and potential therapeutic target for atrial fibril-
120(1), 133–149. lation in old adults. Oncotarget, 7(10), 10803–10811.
9. Wilusz, J. E., Sunwoo, H., & Spector, D. L. (2009). Long non- 31. Qian, C., et al. (2019). Identification of functional lncRNAs in
coding RNAs: Functional surprises from the RNA world. Genes atrial fibrillation by integrative analysis of the lncRNA-mRNA
& Development, 23(13), 1494–1504. network based on competing endogenous RNAs hypothesis. Jour-
10. Luo, X., Yang, B., & Nattel, S. (2015). MicroRNAs and atrial nal of Cellular Physiology, 234(7), 11620–11630.
fibrillation: Mechanisms and translational potential. Nature 32. Lu, J., et al. (2019). Long noncoding RNA GAS5 attenuates car-
Reviews. Cardiology, 12(2), 80–90. diac fibroblast proliferation in atrial fibrillation via repressing
11. Staerk, L., et al. (2017). Atrial fibrillation: Epidemiology, patho- ALK5. European Review for Medical and Pharmacological Sci-
physiology, and clinical outcomes. Circulation Research, 120(9), ences, 23(17), 7605–7610.
1501–1517. 33. Liu, L., Luo, F., & Lei, K. (2021). Exosomes containing
12. Haïssaguerre, M., et al. (1998). Spontaneous initiation of atrial LINC00636 inhibit MAPK1 through the miR-450a-2-3p overex-
fibrillation by ectopic beats originating in the pulmonary veins. pression in human pericardial fluid and improve cardiac fibrosis in
New England Journal of Medicine, 339(10), 659–666. patients with atrial fibrillation. Mediators of Inflammation, 2021,
13. Gramley, F., et  al. (2009). Age-related atrial fibrosis. Age 9960241.
(Dordrecht, Netherlands), 31(1), 27–38. 34. Sun, H., Zhang, J., & Shao, Y. (2021). Integrative analysis reveals
14. Yue, L., et al. (1997). Ionic remodeling underlying action poten- essential mRNA, long non-coding RNA (lncRNA), and circular
tial changes in a canine model of atrial fibrillation. Circulation RNA (circRNA) in paroxysmal and persistent atrial fibrillation
Research, 81(4), 512–525. patients. Anatolian Journal of Cardiology, 25(6), 414–428.
15. Nattel, S., Burstein, B., & Dobrev, D. (2008). Atrial remodeling 35. Dai, H., et al. (2021). LncRNA nuclear-enriched abundant tran-
and atrial fibrillation: Mechanisms and implications. Circulation script 1 regulates atrial fibrosis via the miR-320/NPAS2 axis in
Arrhythmia and Electrophysiology, 1(1), 62–73. atrial fibrillation. Front Pharmacol, 12, 64–7124.
16. Burstein, B., & Nattel, S. (2008). Atrial fibrosis: Mechanisms and 36. Wu, N., et al. (2020). Identification of long non-coding RNA and
clinical relevance in atrial fibrillation. Journal of the American circular RNA expression profiles in atrial fibrillation. Heart, Lung
College of Cardiology, 51(8), 802–809. & Circulation, 29(7), e157–e167.
17. Moran, V. A., Perera, R. J., & Khalil, A. M. (2012). Emerging 37. Zhao, L., et al. (2020). Analysis of long non-coding RNA and
functional and mechanistic paradigms of mammalian long non- mRNA profiles in epicardial adipose tissue of patients with atrial
coding RNAs. Nucleic Acids Research, 40(14), 6391–6400. fibrillation. Biomedicine & Pharmacotherapy, 121, 109634.
18. Guttman, M., et al. (2011). lincRNAs act in the circuitry control- 38. Ke, Z. P., et al. (2019). RNA sequencing profiling reveals key
ling pluripotency and differentiation. Nature, 477(7364), 295–300. mRNAs and long noncoding RNAs in atrial fibrillation. Journal
19. Loewer, S., et al. (2010). Large intergenic non-coding RNA-RoR of Cell Biochemistry, 121, 3752.
modulates reprogramming of human induced pluripotent stem 39. Wu, J., et al. (2019). Identification of atrial fibrillation-associated
cells. Nature Genetics, 42(12), 1113–1117. lncRNAs in atria from patients with rheumatic mitral valve dis-
20. Guttman, M., et al. (2009). Chromatin signature reveals over a ease. Microscopy Research and Technique, 82(7), 1136–1144.
thousand highly conserved large non-coding RNAs in mammals. 40. Wang, Y., et al. (2019). LncRNA NRON alleviates atrial fibrosis
Nature, 458(7235), 223–227. via promoting NFATc3 phosphorylation. Molecular and Cellular
21. Pang, K. C., Frith, M. C., & Mattick, J. S. (2006). Rapid evolution Biochemistry, 457(1–2), 169–177.
of noncoding RNAs: Lack of conservation does not mean lack of 41. Mei, B., et al. (2018). Long non-coding RNA expression profile in
function. Trends in Genetics, 22(1), 1–5. permanent atrial fibrillation patients with rheumatic heart disease.
22. Rinn, J. L., & Chang, H. Y. (2012). Genome regulation by long European Review for Medical and Pharmacological Sciences,
noncoding RNAs. Annual Review of Biochemistry, 81, 145–166. 22(20), 6940–6947.
23. Shi, J., et al. (2021). Circulating long noncoding RNA, GAS5, as 42. Chen, G., et al. (2016). Long non-coding RNA AK055347 is
a novel biomarker for patients with atrial fibrillation. Journal of upregulated in patients with atrial fibrillation and regulates mito-
Clinical Laboratory Analysis, 35(1), 23572. chondrial energy production in myocardiocytes. Molecular Medi-
24. Zeng, W., & Jin, J. (2020). The correlation of serum long non- cine Reports, 14(6), 5311–5317.
coding RNA ANRIL with risk factors, functional outcome, and 43. Ruan, Z., et al. (2015). Long non-coding RNA expression profile
prognosis in atrial fibrillation patients with ischemic stroke. Jour- in atrial fibrillation. International Journal of Clinical and Experi-
nal of Clinical Laboratory Analysis, 34(8), e23352. mental Pathology, 8(7), 8402–8410.
25. Yao, L., et al. (2020). LncRNA MIAT/miR-133a-3p axis regulates 44. Bruneau, B. G. (2013). Signaling and transcriptional networks
atrial fibrillation and atrial fibrillation-induced myocardial fibro- in heart development and regeneration. Cold Spring Harbor Per-
sis. Molecular Biology Reports, 47(4), 2605–2617. spectives in Biology, 5(3), 8292.
26. Ruan, Z. B., et al. (2020). Identification of circulating lncRNA 45. Yeh, Y. H., et al. (2013). Region-specific gene expression profiles
expression profiles in patients with atrial fibrillation. Disease in the left atria of patients with valvular atrial fibrillation. Heart
Markers, 2020, 8872142. Rhythm, 10(3), 383–391.
27. Wang, L. Y., et al. (2019). LncRNA-LINC00472 contributes to the 46. Wijffels, M. C., et al. (1995). Atrial fibrillation begets atrial fibril-
pathogenesis of atrial fibrillation (Af) by reducing expression of lation. A study in awake chronically instrumented goats. Circula-
JP2 and RyR2 via miR-24. Biomed Pharmacother, 120, 109–364. tion, 92(7), 1954–1968.
28. Su, Y., et al. (2018). The long noncoding RNA expression profiles 47. Viereck, J., & Thum, T. (2017). Circulating noncoding RNAs
of paroxysmal atrial fibrillation identified by microarray analysis. as biomarkers of cardiovascular disease and injury. Circulation
Gene, 642, 125–134. Research, 120(2), 381–399.

13

772 Molecular Biotechnology (2022) 64:758–772

48. Tay, Y., Rinn, J., & Pandolfi, P. P. (2014). The multilayered com- 59. Chen, S., et al. (2014). Cardiac miR-133a overexpression prevents
plexity of ceRNA crosstalk and competition. Nature, 505(7483), early cardiac fibrosis in diabetes. Journal of Cellular and Molecu-
344–352. lar Medicine, 18(3), 415–421.
49. Wang, W., et al. (2015). Transcriptome analysis of canine car- 60. Duisters, R. F., et al. (2009). miR-133 and miR-30 regulate con-
diac fat pads: Involvement of two novel long non-coding RNAs nective tissue growth factor: Implications for a role of microRNAs
in atrial fibrillation neural remodeling. Journal of Cellular Bio- in myocardial matrix remodeling. Circulation Research, 104(2),
chemistry, 116(5), 809–821. 170–178.
50. Li, Z., et al. (2017). Altered long non-coding RNA expression 61. Liu, Y., et al. (2021). Identifying ceRNA networks associated
profile in rabbit atria with atrial fibrillation: TCONS_00075467 with the susceptibility and persistence of atrial fibrillation through
modulates atrial electrical remodeling by sponging miR-328 to weighted gene co-expression network analysis. Frontiers in Genet-
regulate CACNA1C. Journal of Molecular and Cellular Cardiol- ics, 12, 653–474.
ogy, 108, 73–85. 62. Sun, F., et al. (2019). LncRNA NRON alleviates atrial fibrosis
51. Lu, Y., et  al. (2010). MicroRNA-328 contributes to adverse through suppression of M1 macrophages activated by atrial myo-
electrical remodeling in atrial fibrillation. Circulation, 122(23), cytes. Bioscience Report, 39(11), BSR20192215.
2378–2387. 63. Li, J., Zhang, Q., & Jiao, H. (2021). LncRNA NRON promotes M2
52. Shen, C., et al. (2018). YY1-induced upregulation of lncRNA macrophage polarization and alleviates atrial fibrosis through sup-
KCNQ1OT1 regulates angiotensin II-induced atrial fibrillation pressing exosomal miR-23a derived from atrial myocytes. Journal
by modulating miR-384b/CACNA1C axis. Biochemical and Bio- of the Formosan Medical Association, 120(7), 1512–1519.
physical Research Communications, 505(1), 134–140. 64. Xuan, L., et al. (2017). Circulating long non-coding RNAs NRON
53. Du, J., et al. (2020). Long noncoding RNA TCONS-00106987 and MHRT as novel predictive biomarkers of heart failure. Jour-
promotes atrial electrical remodelling during atrial fibrillation by nal of Cellular and Molecular Medicine, 21(9), 1803–1814.
sponging miR-26 to regulate KCNJ2. Journal of Cellular and 65. Cao, F., et al. (2019). LncRNA PVT1 regulates atrial fibrosis
Molecular Medicine, 24(21), 12777–12788. via miR-128-3p-SP1-TGF-beta1-Smad axis in atrial fibrillation.
54. Luo, X., et  al. (2013). MicroRNA-26 governs profibrillatory Molecular Medicine, 25(1), 7.
inward-rectifier potassium current changes in atrial fibrillation. 66. Leblanc, F. J. A., et al. (2021). Transcriptomic profiling of canine
The Journal of Clinical Investigation, 123(5), 1939–1951. atrial fibrillation models after one week of sustained arrhythmia.
55. Dai, W., et al. (2020). Long noncoding RNA HOTAIR functions Circulation: Arrhythmia and Electrophysiology, 109, 69.
as a competitive endogenous RNA to regulate connexin43 remod- 67. Mondal, T., et al. (2015). MEG3 long noncoding RNA regulates
eling in atrial fibrillation by sponging microRNA-613. Cardiovas- the TGF-β pathway genes through formation of RNA-DNA triplex
cular Therapeutics, 2020, 5925342. structures. Nature Communications, 6, 7743.
56. Zhao, J. B., et  al. (2018). Modulative effects of lncRNA 68. Harada, M., et al. (2012). Transient receptor potential canonical-3
TCONS_00202959 on autonomic neural function and myocar- channel-dependent fibroblast regulation in atrial fibrillation. Cir-
dial functions in atrial fibrillation rat model. European Review culation, 126(17), 2051–2064.
for Medical and Pharmacological Sciences, 22(24), 8891–8897.
57. Shan, H., et al. (2013). Upregulation of microRNA-1 and micro- Publisher's Note Springer Nature remains neutral with regard to
RNA-133 contributes to arsenic-induced cardiac electrical remod- jurisdictional claims in published maps and institutional affiliations.
eling. International Journal of Cardiology, 167(6), 2798–2805.
58. Li, N., Zhou, H., & Tang, Q. (2018). miR-133: A suppressor of
cardiac remodeling? Frontiers in Pharmacology, 9, 903.

13

You might also like