Next Article in Journal
Structural and Functional Outcomes in Rheumatoid Arthritis After 10-Year Therapy with Disease-Modifying Antirheumatic Drugs Under Tight Control: Evidence from Real-World Cohort Data
Previous Article in Journal
A Retrospective Cohort Study Comparing Robot-Assisted and Conventional Fluoroscopy-Guided Pedicle Screw Placement
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Hidden Regulators: MicroRNAs in Pediatric Heart Development and Disease

by
Adam Kozik
1,†,
Michał Piotrowski
1,†,
Julia Izabela Karpierz
1,2,*,
Mariusz Kowalewski
3 and
Jakub Batko
1,3
1
CAROL—Cardiothoracic Anatomy Research Operative Lab, Department of Cardiovascular Surgery and Transplantology, Institute of Cardiology, Jagiellonian University Medical College, 31-008 Krakow, Poland
2
Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-055 Katowice, Poland
3
Department of Cardiac Surgery and Transplantology, National Medical Institute of the Ministry of the Interior and Administration, 137 Wołoska Street, 02-507 Warsaw, Poland
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
J. Clin. Med. 2025, 14(19), 6833; https://doi.org/10.3390/jcm14196833
Submission received: 31 August 2025 / Revised: 18 September 2025 / Accepted: 23 September 2025 / Published: 26 September 2025
(This article belongs to the Section Cardiology)

Abstract

The development and function of the heart are governed by a highly coordinated network of regulatory mechanisms, among which miRNAs play a central role. These small, non-coding molecules modulate gene expression predominantly through mRNA degradation. This narrative review aims to summarize current knowledge about biogenesis, its impact on heart development and function, and its clinical implications in pediatric cardiology. We discuss how specific miRNAs contribute to shaping the normal heart and influencing the pathogenesis of congenital malformations. Furthermore, we review disease-specific miRNA signatures identified in the most common congenital heart defects and some acquired diseases, including hypoplastic left heart syndrome (HLHS), tetralogy of Fallot (TOF), bicuspid aortic valve (BAV), septation defects, cardiomyopathies, arrhythmias, and myocarditis. Many studies indicate that circulating and tissue miRNAs can become non-invasive biomarkers for early diagnosis and disease monitoring. Experimental data suggest their potential use in treatment despite many delivery and safety challenges. However, further research is necessary to fully exploit the potential of miRNAs and effectively translate these findings into clinical practice in pediatric cardiology.

1. Introduction

MicroRNAs (miRNA) are small, non-coding RNA molecules, typically 19–25 nucleotides in length, that play a crucial role in the post-transcriptional regulation of gene expression. By binding to complementary sequences within target messenger RNAs (mRNAs), they can promote mRNA degradation or inhibit translation, thus influencing a wide range of biological processes. Since their discovery in the early 1990s, miRNAs have emerged as essential regulators in cardiovascular development, physiology, and pathology [1,2]. In recent years, significant progress has been made in understanding their role not only in adult cardiology but also in pediatric cardiac conditions [1].
Pediatric cardiology presents unique challenges compared to adult practice. Many cardiovascular disorders in children are congenital in origin, and their pathophysiology is often linked to disturbances in embryonic cardiac development, genetic syndromes, or early-life environmental influences [1,3]. Even cardiovascular diseases seen in adults, such as myocarditis or cardiomyopathies, can differ significantly in their clinical course and molecular mechanisms from those in children [4,5]. Therefore, biomarkers and therapeutic targets need to be explicitly adapted to the pediatric population. In this context, miRNAs represent a promising tool for diagnosis, prognosis, and potentially treatment of various cardiac diseases in children.
Alterations in miRNA expression patterns have been reported in several types of CHDs, with great heterogeneity between different CHDs and even between treatment stages [6,7,8,9,10,11,12].
Beyond congenital conditions, miRNAs are also involved in pediatric acquired cardiac diseases. For example, distinct circulating miRNA signatures have been reported in children with myocarditis [13,14,15,16,17]. In some cases, these miRNA patterns correlate with disease severity, ventricular function, or treatment response, suggesting their potential utility as non-invasive biomarkers. Furthermore, miRNAs have been implicated in cardiac remodeling, arrhythmogenesis, and heart failure progression in young patients, making them relevant for long-term disease monitoring.
This study is a narrative review that synthesizes current evidence on the miRNAs’ role in diagnosis, treatment, and understanding of various pediatric cardiac conditions. The review does not adhere to a registered protocol and does not employ a systematic search strategy. Instead, articles were selected through targeted searches of major databases (MEDLINE, Embase, Scopus, Google Scholar) and manual review of reference lists, with an emphasis on clinically relevant and recent studies. Consequently, this review may not include all available publications on the subject, and its findings should be interpreted in light of these methodological limitations.

2. Biology of MicroRNA: Mechanisms and Functions

2.1. miRNA Biogenesis

Over the past three decades, since the teams of Victor Ambros and Gary Ruvkin published groundbreaking papers first describing the functions of what was later recognized as encoding miRNAs, our understanding of these small regulatory RNAs has advanced substantially [18,19,20]. Advances in research have allowed the identification of several miRNA biogenesis pathways, which can be broadly classified into canonical and non-canonical [21]. In the canonical pathway, transcription is carried out by RNA polymerase II (Pol II). The primary transcript, known as a pri-miRNA, contains a characteristic hairpin-like structure [22]. This feature is essential for recognition by the Microprocessor complex, which consists of the Drosha endonuclease with RNase III activity and its two partner proteins, DGCR8. The Microprocessor cleaves both strands of the hairpin stem, releasing a precursor molecule called pre-miRNA [23].
This nuclear processing is followed by cytoplasmic maturation. Pre-miRNA is exported via the activity of Exportin 5 and RAN-GTP proteins [24]. Then, the transcript is processed by Dicer—a protein that, like Drosha, possesses RNase III domains, allowing for further cleavage near the loop and the formation of a duplex consisting of the miRNA strand and a complementary “passenger strand” [25]. In the next step, the duplex binds to proteins from the Argonaute (Ago) family, supported by the chaperones HSC70 and HSP90 [26]. One strand is degraded, and the other becomes a fully mature miRNA, approximately 22 nucleotides long. The pU or pA content at the 5’ region determines which of the strands is retained, due to specific interactions with the MID domain of the Ago2 protein [27]. MiRNA bound to Ago proteins forms an RNA-induced silencing complex (RISC), which can recognize and lead to the degradation of target mRNAs [28]. Most miRNAs are produced as a result of the process described above, but there are also several non-canonical synthesis pathways that bypass some of the steps or utilize unusual precursors (Figure 1).
In the canonical pathway, after transcription by Pol II, the pri-miRNA forms a hairpin-like structure, which is cleaved by the Microprocessor. The released pre-miRNA is transported from the nucleus to the cytosol by Exportin 5 and RAN-GTP. After cleavage by Dicer, a ~22 bp miRNA duplex is formed, and one of the strands is degraded. The mature miRNA, when bound to the Argonaute protein (Ago), forms a silencing complex (RISC). In the less common non-canonical pathways, the requirement for Microprocessor or Dicer cleavage can be bypassed. In the case of so-called mirtrons, excision of an intron by the spliceosome directly generates a pre-miRNA. The transcript bypasses the Microprocessor complex and is immediately exported to the cytoplasm, where it undergoes further processing by Dicer. In contrast, miRNA-451 maturation requires Drosha, but not Dicer activity. Its nuclear processing is similar to that of canonical miRNAs. However, once exported to the cytoplasm, pre-miRNA-451 binds directly to Ago, whose catalytic activity generates the mature miRNA. Upon recognition of a target mRNA, the RISC can trigger its degradation through the action of the exonuclease XRN-1. Post-transcriptional inhibition may also occur through interactions of the RISC with the ribosome.
For example, mirtrons produced by transcription of some introns are immediately exported to the cytosol, bypassing cleavage by Drosha [29]. Other miRNAs, such as miR-451, are Dicer-independent, and the final product is formed by cleavage of the 3’ arm of the hairpin by Argo2 protein [30]. Understanding the subsequent stages of transcript maturation opens new possibilities for the development of synthetic miRNAs with therapeutic potential.

2.2. Regulation of miRNA Expression and Decay

Most miRNAs originate from introns or non-coding transcripts, while a minor fraction overlaps with exons of protein-coding genes [21]. Their expression can be regulated at multiple levels, including transcription, maturation, or degradation of the final product. Pol II regulates the transcription process in a manner analogous to that of protein-coding genes for all canonical and most non-canonical miRNAs [31]. In addition, numerous mechanisms modulate the activity of pivotal enzymes such as Drosha or Dicer [32]. Post-transcriptional modifications of pri- or pre-miRNAs represent another essential layer of regulation. One of the best-characterized examples is Lin-28-mediated oligo(U) tailing of the let-7 family, which blocks Dicer processing [33]. Notably, even single-nucleotide changes can sometimes be crucial. For instance, 3′-end monouridylation exerts the opposite effect, enhancing maturation [34]. Likewise, adenosine deamination can trigger pri-miRNA degradation by adenosine deaminases acting on RNA (ADARs) [35]. Additionally, many RNA-binding proteins, such as hnRNP A1 or SRSF1, can regulate the activity of specific miRNAs through diverse mechanisms [36].
The biological functions of miRNAs are mainly dependent on their stability. Most have biological half-lives of several hours, but there are many exceptions [37]. For example, in retinal photoreceptor cells, the levels of many miRNAs change dynamically in response to light stimuli [38,39]. One of the key mechanisms responsible for this phenomenon is target-directed miRNA degradation (TDMD). The binding of an mRNA to sequences at the miRNA 3′ end, distinct from classical miRNA-RNA interactions that typically involve the seed region at the miRNA 5′ end, triggers the recruitment of the ZSWIM8 ubiquitin ligase. This, in turn, promotes miRNA ubiquitination and degradation via the action of specific exonucleases [40].

2.3. Mechanism of Action

Although Ago proteins within the RISC possess exoribonucleolytic activity and contain a PIWI domain with a catalytic center resembling RNase H, degradation of target mRNAs is rarely the result of direct cleavage [41,42,43]. Upon mRNA binding to the RISC, additional proteins such as TNRC6 are recruited. Their interaction with the poly(A)-binding protein (PABPC) leads to the activation of protein complexes with deadenylase activity, among which the CCR4-NOT complex plays a central role [44,45]. Poly(A) tail shortening weakens PABPC interactions with the translation initiation factors eIF4E and eIF4G, exposing the 5′ cap structure to the Dcp2 decapping enzyme. Removal of the cap renders the transcript susceptible to 5′→3′ degradation by the Xrn1 exonuclease [46]. While mRNA destabilization is the primary mechanism responsible for miRNA-mediated repression, interactions between RISC components and specific protein partners can also directly inhibit translation [47].
Target recognition by miRNAs most often relies on binding sites that promote TNRC6-dependent repression and span nucleotides 2–7 of the miRNA (the so-called seed region), usually extended by two additional nucleotides, and located within the 3′UTR of the mRNA [48]. In some cases, 3′-compensatory sites can also be utilized, which bind to extensive sequences at the 3′ end of the mRNA in the case of an incomplete seed region match. Other non-canonical binding sites have also been described; however, these typically involve short stretches of complementarity, and seed region pairing remains essential for efficient translational repression [21].

3. Role of miRNA in Cardiac Development and Physiology

MiRNA, by regulating gene expression, plays a key role in prenatal development [49]. In recent years, its role in cardiogenesis and its influence on the occurrence of heart defects have been increasingly emphasized [50]. For example, deletion of the Dicer enzyme in mice, which is essential for the maturation of canonical miRNAs, led to disruption of heart septation and prenatal death [51]. In turn, its cardiac-restricted deletion in 3-week-old mice resulted in the development of lethal dilated cardiomyopathy [52]. This suggests that miRNA is essential not only for cardiogenesis but also for the maturation and proper functioning of this organ [2]. Moreover, changes in miRNA expression in many cardiovascular diseases indicate their role in maintaining adequate homeostasis and provide hope for the development of new therapies and diagnostic methods.
To better understand the role of miRNA in cardiogenesis, it is worth emphasizing that its levels vary between different tissues. It is estimated that the majority, as much as 45%, of miRNAs in the rodent heart are miR-1 [53]. Several other miRNAs, termed MyomiRs, have also been identified in large numbers in the developing heart. These include miR-133 and miR-206, miR-143 and miR-208 [50]. The functions of some key miRNAs involved in development are summarized in Table 1.

3.1. miRNA as a Regulator of Cardiomyocyte Maturation

A fundamental role in cardiogenesis is played by the specification of cardiac mesoderm under the influence of growth factors such as bone morphogenetic protein (BMP), fibroblast growth factor (FGF), and wingless-type (WNT) protein family [50]. Central to this process are miR-1 and miR-133, which originate from two genomic clusters: miR-1/133a-1 on chromosome 18 and miR-1/133a-2 on chromosome 20 [54].
MiR-133 along with miR-499 influences the expression of transcription factors such as GATA4 and Nkx2.5, and cardiac proteins like connexin 43 (Cx43) and cardiac troponin T (cTnT), with both miRNAs exhibiting synergistic effects [55].
Moreover, miR-499 plays an essential role in remodeling type II (fast) muscle fibers toward type I (slow) fibers. Along with miR-208b, encoded in the intron of the myosin heavy chain gene, it is critical for modulating its expression by suppressing transcriptional repressors such as Sox6, Purβ, and Sp3 [56].
A promising potential biomarker is miR-20b-5p, acting as an inhibitor of cardiac cell differentiation [57]. Its elevated levels have been observed in patients with atrial septal defect (ASD). Its mechanism involves inhibition of transcription factors such as the GATA4 above and Nkx2.5 via repression of Tet methylcytosine dioxygenase 2 (TET2)—an enzyme catalyzing epigenetic modifications that promote transcription [57]. Alongside this, miR-128a plays a regulatory role in heart development, modulating the differentiation of cardiac precursor cell populations. It likely acts as a switch that determines the direction of differentiation of pluripotent cells—whether they become cardiomyocytes or cells of the conduction system [58].

3.2. miRNA as a Regulator of Proliferation and Cell Cycle

Alongside miR-1, one of the most critical proliferation inhibitors is the miR-15 family. Expression of miR-15a and miR-16-1 increases between days 1 and 9 after birth in mice, leading to the inhibition of a broad spectrum of cell cycle-related genes such as Chek1, Cdc25A, Ccnd1, and Ccne1 [59].
In contrast, the miR-302-367 cluster, highly expressed in embryonic stem cells, promotes cell division and is essential for maintaining pluripotency [60]. It acts similarly to the miR-17-92 cluster, which inhibits key genes for cell cycle progression such as Cdkn1a, Rb1, and E2F1, and whose function is essential even during postnatal development [61]. MiR-410 and miR-495 also play an important role in stimulating cardiomyocyte proliferation at the prenatal stage. Both act by targeting the transcriptional coactivator Cited2 [62]. Another factor supporting proliferation is the miR-106b~25 cluster, which is highly expressed in mice during the first two weeks after birth. It inhibits numerous cell cycle inhibitors, such as Cdkn1a and Wee1. Loss of its function manifested as mild but significant eccentric hypertrophy and reduced ejection fraction, suggesting this cluster maintains the balance between cardiomyocyte hypertrophy and hyperplasia [63]. Additionally, a high-throughput functional screening study identified miR-515-3p and miR-519e-3p as potent inducers of proliferation in cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs) [64].

3.3. miRNA as a Regulator of Angiogenesis

Adequate vascularization of the myocardium plays a critical role in supporting its regeneration after infarction, making the identification of miRNAs influencing this process crucial for developing new therapeutic strategies. One of the most important promoters of angiogenesis is miR-126, which enhances the endothelial response to VEGF by suppressing endogenous inhibitors of this pathway (SPRED1 and PIK3R2) by directly inhibiting their translation [65]. In response to hypoxia, miR-210, which plays a similar role, is activated [66]. Conversely, miR-92a acts as an inhibitor, and its suppression promotes neovascularization following myocardial ischemia [67]. This insight led to the development of a synthetic microRNA-92a inhibitor (MRG-110) aimed at promoting wound healing in both diabetic and non-diabetic wounds [68]. Notably, a phase I clinical trial evaluating the safety of MRG-110 following intradermal administration in healthy volunteers was completed in 2019 (NCT03603431). Another significant cluster is miR-143/145. It regulates the ability of VSMCs to adopt a communicative contractile phenotype by suppressing ACE expression. Their loss leads to chronic activation of the RAA system, contractile dysfunction, and the initiation of neointimal changes, making them a promising target for the treatment of vascular damage [69].

3.4. miRNA as a Regulator of the Cardiac Conduction System

The cardiac conduction system (CCS) is crucial for the generation and conduction of impulses that initiate myocardial contraction. Its most important components include the sinoatrial node (SAN), which serves as the primary pacemaker, the atrioventricular node, the His bundle, its branches, and a network of Purkinje fibers [70]. miRNAs play essential roles in their development. Deletion of miR-17-92 and miR-106b-25 was found to lead to increased expression of the Shox2 and Tbx3 genes, key players in SAN development. Loss of function of these clusters significantly increased the risk of developing atrial fibrillation (AF) in mice [71]. Many miRNAs also play a role in regulating the function of specific ion channels, e.g., HCN (hyperpolarization-activated cyclic nucleotide-gated), which are crucial for the pacemaker function of SAN [72]. miR-486-3p has been shown to inhibit HCN4 expression, leading to a decrease in heart rate. The same receptor is also acted on by miR-370-3p, whose silencing in heart failure can restore normal pacemaker function. [73]. Furthermore, miR-423-5p regulates HCN4 in the context of bradycardia induced by regular sports training [74]. The previously mentioned miR-1 also influences the expression of KCNJ2, KCND2, and GJA1 channels, and together with miR-133 regulates calcium cycling by modulating ryanodine receptor (RyR2) function [2,75,76]. Additionally, miR-208a is crucial for the expression of some connexins, proteins necessary for proper conductance [77].
Table 1. Function of selected miRNAs in cardiac development.
Table 1. Function of selected miRNAs in cardiac development.
microRNAPrimary FunctionTargeted Genes/PathwaysEffect of DysregulationReferences
miR-1Inhibition of the cell cycle Myocardin, WNT, FGFKnockout: dilated cardiomyopathy due to sarcomere disruptions and postnatal lethality (murine)[78,79]
miR-133Anti-apoptotic protection and synergistic effects with miR-499GATA4, Nkx2.5, Cx43, cTnTKnockout: dilated cardiomyopathy, VSD, fibrosis (murine)[55]
miR-499Muscle fiber remodeling (type II→type I) and cardiomyocyte differentiationSox6, Purβ, Sp3, GATA4, Nkx2.5, Cx43, cTnTOverexpression: conversion of muscle fibers to slow type (murine)[55,56]
miR-208bMuscle fiber remodeling and myosin heavy chain expressionSox6, Purβ, Sp3, myosin heavy chain geneKnockout: inhibition of the formation of slow muscle fibers (murine)[56,80]
miR-20b-5pInhibition of cardiac cell differentiationTET2, GATA4, Nkx2.5Knockout: more cTnT-positive cells
Overexpression: fewer cTnT-positive cells (hESCs-CMs)
[57]
miR-128aRegulation of differentiation, guidance toward a cardiomyocyte or conduction system phenotypeCardiac precursor cell differentiation pathwaysKnockout: slower heart rate, diminished ventricular size (zebrafish)[58]
miR-15aInhibition of proliferation Chek1, Cdc25A, Ccnd1, Ccne1Knockout: increased number of cells entering mitosis, with cell cycle arrest (murine)[59]
miR-16-1
miR-17-92Progression of the cell cycleCdkn1a, Rb1, E2F1[61]
miR-106b~25Regulation of the hyperplasia–hypertrophy balanceE2f5, Cdkn1c, Ccne1, Wee1, Hand2, Mef2dKnockout: mild eccentric hypertrophy and reduced ejection fraction (murine)
Overexpression: hyperplasia, increased proliferation (murine)
[63]

4. Clinical Applications of MicroRNA in Pediatric Cardiology

4.1. miRNA as Potential Biomarkers for Congenital Heart Disease

Multiple studies aiming to identify potential prenatal biomarkers of congenital heart diseases (CHDs) were conducted in recent years as a possible solution for the limitations of prenatal ultrasound [17,81,82,83,84]. As stated before, miRNAs play a crucial role in the development of the cardiovascular system, and alterations in their expression patterns have been implicated in the pathogenesis of congenital heart diseases. Several studies have explored whether specific miRNA profiles in maternal or fetal blood could serve as non-invasive biomarkers for prenatal CHD detection. Zhu et al. reported that circulating miR-19b, miR-22, miR-29c, and miR-375 were significantly elevated in pregnant women carrying fetuses with CHD, achieving good diagnostic accuracy [85]. Similarly, Gu et al. identified a panel including miR-142-5p, miR-1275, miR-4666a-3p, and miR-3664-3p that effectively distinguished CHD cases from healthy controls [86]. Beyond diagnostic associations, mechanistic insights were provided by studies on miR-29b-3p, which was found to be increased in the right ventricular outflow tract of CHD patients [87]. Experimental overexpression of this miRNA in zebrafish embryos caused developmental delay, cardiac malformations, and impaired function, while its inhibition promoted cardiomyocyte proliferation. This effect was shown to involve direct targeting of NOTCH2, a pathway relevant to cardiac development. Kong et al. demonstrated that miR-1, miR-208, and miR-499 were reduced in umbilical cord blood from CHD fetuses, with distinct expression trends depending on the CHD subtype. These miRNAs showed good discriminatory potential for differentiating affected from healthy fetuses [88]. Other studies primarily focused on differences in miRNA expression patterns without directly assessing their diagnostic performance. For example, Yang et al. reported upregulation of miR-1-3p, miR-184, and miR-206, while Kehler et al. described increased expression of miR-99a [81,89]. Similarly, Jin et al. and others identified additional miRNAs associated with CHD [84]. Collectively, these findings further support the concept that circulating miRNAs may serve as clinically relevant biomarkers, even though their diagnostic potential remains to be fully explored. The mentioned studies suggest that specific miRNAs—such as miR-29b-3p, miR-1, miR-208, and miR-499—may serve as promising non-invasive biomarkers for the early detection of CHD, with potential applications in prenatal screening programs. Its clinical significance and possibility to introduce it into everyday practice are still to be investigated in the future before the development of specific test panels. Current literature data indicate that miRNAs are promising biomarkers for congenital heart diseases. However, significant heterogeneity across published studies must be taken into account. Differences in sample sizes, patient populations, and analytical methods hinder their clinical applicability [90]. It is worth noting that the studies cited above examined various types of materials, such as maternal or umbilical blood or heart tissue samples [85,86,87,88]. These limitations highlight the need for larger, standardized, multicenter investigations before clinical implementation can be considered.

4.2. miRNAs in Congenital Heart Diseases

CHDs were traditionally classified into cyanotic and non-cyanotic forms, depending on whether the defect leads to mixing of oxygenated and deoxygenated blood with resultant systemic desaturation [91,92]. Common cyanotic lesions include TOF, transposition of the great arteries (TGA), truncus arteriosus (TA), and total anomalous pulmonary venous return. In contrast, typical non-cyanotic CHDs include ventricular septal defect (VSD), atrial septal defect (ASD), patent ductus arteriosus (PDA), and coarctation of the aorta (CoA). Beyond differences in clinical presentation, prognosis, and therapeutic strategies, emerging evidence suggests that cyanotic and non-cyanotic CHDs also exhibit distinct molecular signatures, including differential miRNA expression.
In one study, the expression levels of miR-21-5p, miR-15-5p, miR-221-3p, and miR-26-5p were found to differ between children with CHD and healthy controls significantly. Notably, miR-21-5p levels were particularly elevated in patients with cyanotic CHD compared to those with non-cyanotic defects. These findings suggest that specific miRNA dysregulation may contribute not only to the pathogenesis of cardiac malformations but also to the variable severity observed across CHD subtypes. The pronounced upregulation of miR-21-5p in cyanotic CHD, for instance, may reflect its association with greater disease burden and adverse clinical course [93].
More recent classification systems take into account hemodynamic and anatomical complexities, providing a framework that is more clinically relevant. To date, no studies have examined miRNA profiles in the context of these modern classification systems.
Further studies concerning those differences, especially during prenatal time, might be beneficial for early detection of those conditions and prediction of their severity. Below, we discussed current knowledge about the molecular profiles of certain CHDs [93].

4.2.1. Hypoplastic Left Heart Syndrome

Univentricular heart is one of the most severe forms of congenital heart disease, characterized by underdevelopment of the cardiac structures and functional dependence on one ventricle [94]. HLHS is considered one of the deadliest of those conditions [95].
Several studies have explored circulating miRNAs as potential biomarkers of heart failure in patients with univentricular hearts. Abu-Halima and colleagues analyzed blood samples from 48 patients and 32 controls using a large panel of miRNA arrays, identifying 50 significantly dysregulated miRNAs [7]. Among these, miR-150-5p demonstrated the strongest association with heart failure severity, emerging as an independent predictor of overt heart failure. Importantly, its diagnostic performance was comparable to established clinical parameters such as NT-proBNP levels and NYHA functional class, and when combined with NT-proBNP, predictive accuracy was further enhanced [7]. In a complementary line of investigation, Ramachandran and colleagues focused on children with univentricular hearts, assessing whether circulating microvesicle-derived miRNAs could provide insights into heart failure progression [8]. Their findings pointed to miR-129-5p, which was inversely correlated with heart failure severity as measured by the Ross score. Experimental validation in cardiomyocyte models further suggested a mechanistic link between oxidative stress, miR-129-5p downregulation, and altered expression of bone morphogenetic protein receptor 2, a pathway implicated in pulmonary vascular disease [8]. Together, these studies highlight that distinct miRNAs—miR-150-5p and miR-129-5p—hold promise as non-invasive biomarkers in univentricular hearts, potentially offering additive value to traditional measures for risk stratification and disease monitoring. While Abu-Halima emphasized their prognostic utility in predicting overt heart failure, Ramachandran provided mechanistic insights linking miRNA regulation to stress responses in pediatric cardiomyocytes. These complementary perspectives underscore the potential of miRNA profiling to refine biomarker strategies in HLHS.
Beyond circulating biomarkers, studies of myocardial tissue have provided complementary insights into the role of miRNAs in univentricular physiology. Suharov and colleagues examined miRNA expression directly in right ventricle (RV) tissue from children with HLHS and demonstrated that the miRNA profile in this setting was essentially distinct from that observed in adult idiopathic dilated cardiomyopathy, underscoring the uniqueness of developmental mechanisms in congenital heart disease [9]. Dysregulated miRNAs were found to target several transcription factors and signaling pathways fundamental for cardiac morphogenesis, with inverse patterns between miRNA levels and the expression of their predicted targets [9]. Notably, the myocardial miRNA signature was not static but evolved across the stages of surgical palliation. Analysis of the miRNA expression in patients with HLHS revealed significant differences related to right ventricular volume overload. The most noteworthy finding was a normalization of miR-100, miR-99a, and miR-145 levels in patients after stage 3 of surgical palliation (following the Fontan procedure) compared to those without surgery or after stage 1 (the Norwood procedure), when RV volume overload is significant. These dynamic changes suggest that the reduction in RV volume load achieved later in palliation can partially reverse miRNA dysregulation. In contrast, the expression of miR-204 and miR-137-3p remained altered regardless of the stage of surgical treatment. Changes in miR-204 expression were also observed in other patients with idiopathic dilated cardiomyopathy, which may indicate its potential use in pediatric heart failure regardless of etiology [9]. Predicted targets included key transcriptional regulators such as QKI, CDK6, SOX11, FOG2, GATA6, GATA4, and dHAND, several of which showed stage-dependent expression shifts. Together, these findings indicate that some miRNA alterations reflect adaptive mechanisms in response to hemodynamic stress, while others may be linked to intrinsic defects in cardiac development [9].
As HLHS is one of the most fatal and complex CHDs, a better understanding of its pathophysiology is needed to possibly identify prenatal biomarkers and investigate whether specific miRNA profiles are related to hemodynamic and clinical outcomes of surgical treatment of the disease.

4.2.2. Tetralogy of Fallot

Among cyanotic congenital heart diseases, TOF occupies a central role, representing the most frequent lesion within this group. Epidemiological studies indicate that TOF constitutes roughly 5–7% of all congenital cardiac malformations. The condition arises from an abnormal conotruncal development of the heart and is classically described by four interrelated anatomic abnormalities: an anteriorly malaligned ventricular septal defect, overriding of the aortic root, obstruction of the right ventricular outflow tract (subpulmonary and/or valvular), and secondary hypertrophy of the right ventricle. The clinical spectrum is heterogeneous. In the neonatal period, the extent of right ventricular outflow tract obstruction largely determines symptomatology, with severe forms manifesting as profound cyanosis shortly after birth, while milder cases may present later in infancy. Importantly, the hypertrophic response of the right ventricle is not a primary malformation but a physiological consequence of chronic pressure overload [96].
Modern diagnostic pathways have shifted substantially with the introduction of fetal echocardiography, which allows TOF to be recognized before birth and facilitates parental counseling as well as delivery planning in tertiary care centers. In postnatal life, prompt recognition of critical right ventricular outflow tract obstruction and stabilization of affected neonates remain essential to avert hypoxemic crises and cardiovascular decompensation [97]. Current perspectives on management emphasize not only the technical aspects of surgical and catheter-based interventions but also the role of coordinated, multidisciplinary care teams. Optimal outcomes are increasingly linked to timely diagnosis, individualized treatment planning, and effective interprofessional collaboration. Because of the relatively high incidence of TOF compared to other cyanotic CHDs, the miRNA profiles in that condition were thoroughly investigated. For example, Zhu and colleagues found that, in particular maternal circulating miR-22 was upregulated in the case of TOF [85].
One of the earliest comprehensive efforts to profile non-coding RNA signatures in TOF myocardium was carried out by O’Brien and colleagues, who examined RV tissue from infants with nonsyndromic TOF compared with both fetal and typically developing hearts [98]. Their analysis revealed marked changes in both miRNAs and snoRNAs, with expression patterns in TOF myocardium resembling those of fetal samples, suggesting a persistence of developmental transcriptional programs. A subset of 44 genes essential to cardiac development showed altered expression, correlating inversely with 33 deregulated miRNAs. These findings not only highlight potential regulatory networks disrupted in TOF but also point toward aberrant RNA processing, as evidenced by the association between snoRNA dysregulation, spliceosomal variants, and heart development. Expanding on this line, Bittel and colleagues placed particular focus on miR-421, which was significantly upregulated in TOF myocardium and negatively correlated with SOX4, a Notch pathway regulator [99]. Functional experiments confirmed its capacity to influence cardiomyocyte proliferation, positioning miR-421 as a candidate mediator of RV remodeling in TOF. Independent work by Zhang and colleagues identified 18 miRNAs with significantly altered expression in RVOT tissue, among them miR-424 and miR-222, both of which were experimentally shown to modulate cardiomyocyte proliferation and differentiation [100]. Similarly, Liang and colleagues identified miR-940 as one of the most profoundly downregulated miRNAs in TOF myocardium, particularly enriched in the normal RVOT. Functional assays demonstrated that miR-940 loss enhanced progenitor proliferation while impairing migration via direct regulation of JARID2, thus linking altered secondary heart field progenitor dynamics with TOF development [101]. Other groups have focused on well-characterized cardiac miRNAs [102,103]. Wu reported that miR-1 and miR-206 were downregulated in TOF myocardium, resulting in increased expression of connexin-43, a protein crucial for myocardial conduction [102]. Grunert further integrated genome-wide miRNA and mRNA datasets in TOF myocardium, identifying key regulatory axes including miR-1/miR-133 pairs acting on structural and electrophysiological genes such as KCNJ2 and TNNI1 [103]. Collectively, these studies suggest that while many miRNAs in TOF reflect fetal-like transcriptional states, others directly perturb pathways critical to RV proliferation, conduction, and morphogenesis.
The contribution of miRNAs is not restricted to myocardial tissue. Circulating miRNA profiles in patients late after surgical repair TOF repair were systematically explored by Abu-Halima, who reported broad dysregulation across dozens of miRNAs, with several (miR-181d-5p, miR-206, miR-625-5p) showing excellent discriminatory value between TOF patients and controls [6]. Interestingly, miR-421 and miR-625-5p were reduced explicitly in individuals who developed symptomatic right heart failure, underscoring the potential prognostic role of circulating miRNAs in disease progression [6].
Complementary to this, Gomez and colleagues identified increased expression of miR-221-5p, miR-21-5p, and miR-155-5p in children with TOF [104]. Target predictions implicated these miRNAs in hypoxia-sensitive signaling pathways, including AKT, SMAD, and TNF-α, supporting the concept that circulating miRNAs reflect both developmental abnormalities and secondary remodeling processes. More recently, Yang and colleagues analyzed exosomal miRNAs in amniotic fluid from fetuses diagnosed with TOF [105]. Over 250 miRNAs were differentially expressed, with enrichment in Notch and Wnt signaling pathways. Functional experiments highlighted miR-10a-5p as a suppressor of TBX5, a master transcription factor in cardiogenesis, thus providing a possible mechanistic link between prenatal exosomal signaling and impaired cardiomyocyte differentiation. Beyond tissue and circulating profiles, genomic approaches have expanded the picture of miRNA involvement in TOF. Bassett and colleagues analyzed the role of copy number variants (CNVs) overlapping miRNA loci, demonstrating an enrichment of rare CNVs in TOF patients independent of the classic 22q11.2 deletion [106]. This supports a multi-hit model in which structural variation affecting miRNA dosage may contribute to susceptibility. At the systems level, integrative bioinformatics analyses by You and colleagues constructed miRNA–mRNA–TF co-regulatory networks, identifying hub genes and pathways spanning ubiquitin-mediated proteolysis, ribosomal function, and energy metabolism [107]. Such work provides a higher-order view of how diverse miRNA alterations converge on essential developmental and metabolic processes in TOF. Adding another layer, Wang and colleagues applied high-throughput sequencing and uncovered sex-specific differences in small RNA expression, with miR-1/133 accounting for the most significant variance between male and female TOF hearts [108]. This observation raises the possibility that miRNA regulation may partly explain reported sex differences in CHD prevalence and outcomes.

4.2.3. Septation Defects

VSD and ASD are the two most common CHDs, with incidences of 0.03% of newborns in the case of VSD and 0.016% in the case of ASD [109,110]. Prenatal diagnosis of ASD and VSD was investigated by Zhu and colleagues and discussed beforehand.
Ventricular Septal Defect
VSD accounts for nearly 40% of all congenital cardiac malformations and frequently requires surgical intervention [110]. While their etiology is multifactorial, encompassing both genetic and environmental influences, accumulating evidence points to an essential role of miRNAs in their pathogenesis. In an early investigation, Li and colleagues analyzed 25 candidate miRNAs in cardiac tissue from patients with congenital heart disease and healthy controls. They reported that miR-1-1 and miR-181c exhibited disease-specific alterations in individuals with VSD. Reduced miR-1-1 levels were associated with increased expression of GJA1 and SOX9, while elevated miR-181c correlated with downregulation of BMPR2. Functional validation confirmed direct interactions between these miRNAs and their predicted targets, implicating them in septal morphogenesis and suggesting a mechanistic contribution to VSD development [10]. The same group later extended their work to circulating miRNAs. In plasma samples from children with VSD, they identified 36 dysregulated miRNAs, of which RT-qPCR validated eight. Bioinformatic analysis pointed to regulatory networks involving key developmental genes such as NOTCH1, HAND1, ZFPM2, and GATA3. This study provided the first evidence that peripheral miRNA profiles reflect underlying cardiac developmental disturbances, and highlighted their potential as non-invasive biomarkers for VSD [111]. The clinical translation of these findings was further advanced by Gu et al., who investigated maternal serum from pregnancies affected by congenital heart disease. They identified 38 dysregulated circulating miRNAs, with 12 validated by real-time PCR. A biomarker panel consisting of four miRNAs—miR-142-5p, miR-1275, miR-4666a-3p, and miR-3664-3p—demonstrated excellent diagnostic accuracy, achieving an AUC of 0.92 for distinguishing affected pregnancies from controls. Significantly, several miRNAs declined rapidly after delivery, confirming their fetal origin and supporting their utility as non-invasive prenatal diagnostic tools [86]. More recently, attention has shifted to exosome-derived miRNAs, which may provide more stable and specific biomarkers. Jin et al. profiled maternal circulating exosomal miRNAs in pregnancies with fetal VSD and reported 77 differentially expressed candidates. Five were validated, among which hsa-miR-146a-5p emerged as an exceptionally robust biomarker, achieving near-perfect diagnostic performance with an AUC of 0.997 [84].
Atrial Septal Defect
Research into the role of miRNAs in ASD has gradually evolved from genetic association studies to functional and mechanistic insights. The first evidence came from genetic analyses. Yu and colleagues identified a single-nucleotide polymorphism in miR-196a2 (rs11614913 T>C) that was associated with ASD susceptibility [112]. Carriers of the C allele demonstrated a reduced risk of developing ASD, and functional assays suggested that the variant modulated miRNA expression and its influence on cell differentiation and morphogenesis. In the same year, Wang and colleagues described a familial form of ostium secundum ASD II linked to a mutation in the 3′UTR of the ACTC1 gene (c.*1784T>C) [113]. This variant created a de novo binding site for miR-139-5p, leading to pathological repression of ACTC1. Significantly, the effect could be intensified by miR-139-5p mimics and alleviated with inhibitors, demonstrating a direct gain-of-function mechanism where altered miRNA–mRNA interaction contributes to septal malformation.
A shift towards circulating biomarkers followed. Song and colleagues reported that children with ASD had elevated levels of hsa-let-7a, hsa-let-7b, and hsa-miR-486 [11]. Among these, let-7a and let-7b were specific to ASD compared to other septal defects. Remarkably, the same miRNA changes were detectable in the mothers of affected children, suggesting potential use in prenatal screening. Receiver-operating characteristic analyses confirmed their diagnostic accuracy, highlighting circulating and maternal miRNAs as promising non-invasive biomarkers for ASD. At the tissue level, Han and colleagues performed a systematic profiling of atrial septa from ASD fetuses [114]. They identified 70 dysregulated miRNAs, with consistent upregulation of the miR-29 and miR-143/145 clusters, and downregulation of the miR-17-92, miR-106b-25, and miR-503/424 clusters. Mouse model validation showed that all except miR-143/145 were functionally associated with septal defects, pointing to cluster-level dysregulation as a driver of abnormal atrial septation.
Most recently, mechanistic insights into transcriptional regulation have emerged. Li et al. studied a family with a novel splicing mutation in NKX2-5 (c.335-1G>A) [115]. Patient-derived hiPSC cardiomyocytes showed that NKX2-5 haploinsufficiency disrupted apoptosis and proliferation during cardiomyocyte development. Crucially, these effects were mediated through the regulation of miR-19a/b, positioning miRNAs as central downstream effectors of transcription factor dysfunction in ASD [12].
Atrioventricular Defects
On the other hand, atrioventricular defects are relatively less common, constituting from 3 to 7 percent of CHDs. Those defects are often seen in patients with Down Syndrome—40 to 50% of them have this type of septation defect. Therefore, it was proven that miRNAs strictly linked to Down syndrome (found on chromosome 21) are expressed in cardiac tissue of patients with atrioventricular defect [116]. Among them, the miR-99a/let-7c cluster appears to play a key role. MiRNAs within this cluster influence cell differentiation in the early stages of cardiogenesis. Increased levels of miR-99a/let-7c and decreased expression of its targets, such as Ezh2 and Smarca5, have been observed in the hearts of human fetuses with trisomy 21 [117]. Moreover, in a study aimed at identifying genes and miRNAs associated with AVSD in patients with Down syndrome, Wang L et al. identified several miRNAs (miR-518a, miR-518e, miR-518f, miR-528a, and miR-96) that may contribute to the development of AVSD by regulating the expression of genes important for cardiomyocyte differentiation and adhesion (AUTS2 and KIAA2022) [118].

4.2.4. Pulmonary Hypertension Secondary to Congenital Heart Disease

Pulmonary hypertension (PH) is related to a significant medical burden; it is relatively common in cases of some CHDs. Multiple studies investigating the miRNA role in secondary PH were conducted in recent years. Recent studies have highlighted the role of miRNAs as regulators, biomarkers, and potential therapeutic targets. High-throughput sequencing and bioinformatic analyses have revealed disease-specific miRNA signatures in pediatric PH, which not only correlate with invasive hemodynamic indices but also change dynamically during treatment. Circulating molecules such as miR-122-5p, miR-124-3p, miR-204-5p, and miR-9-5p have been shown to decrease with therapy, linking them to key pathways including TGF-β, VEGF, PI3K/Akt, cGMP-PKG, and HIF-1 signaling [119].
Regional profiling of miRNA gradients across the right ventricle and pulmonary circulation provided additional insight into their spatial regulation. In children with pulmonary arterial hypertension (PAH), distinct step-up and step-down patterns of miRNAs such as miR-193a-5p and miR-423-5p were observed, while others, including miR-29a-3p, miR-26a-5p, and miR-200c-3p, were upregulated compared with controls. These expression changes correlated with prognostic parameters such as pulmonary vascular resistance and RV function, suggesting that miRNA gradients may represent a novel layer of epigenetic regulation and biomarker development [120]. Several candidate miRNAs have been identified in CHD-associated PAH specifically. Elevated serum miR-27b and reduced miR-451 were independently associated with increased pulmonary pressures and biochemical markers such as BNP and ADMA, highlighting their potential combined diagnostic utility [121]. Similarly, increased circulating miR-19a distinguished CHD patients with PAH from those without, with good diagnostic accuracy, supporting its role as a clinically useful biomarker [83].
Mechanistic studies further underscore the importance of specific clusters. The miR-143/145 axis has been shown to regulate pulmonary artery smooth muscle cell migration and apoptosis, with miR-143-3p enriched in exosomes and exerting paracrine effects on endothelial cells. Inhibition of miR-143-3p ameliorated experimental PH, confirming its pathogenic role [122]. Other work demonstrated that downregulation of miR-98 in CHD-PAH correlates with pulmonary artery pressures and disease severity. As miR-98 exerts anti-hypertrophic effects in cardiomyocytes, its reduced expression may contribute to vascular remodeling, and ROC analysis indicated strong diagnostic potential [123].
Beyond individual miRNAs, circRNA-miRNA-mRNA networks are beginning to be explored. In pediatric CHD-PAH, specific circRNAs were found to be dysregulated, predicted to target miRNAs implicated in oxidative phosphorylation and endothelial junction biology, suggesting a complex regulatory axis in pulmonary vascular remodeling [124]. Lung tissue analyses confirmed broad alterations in miRNA expression, with miR-27b notably upregulated and linked to NOTCH1 regulation, further connecting it with the pathogenesis of CHD-associated PAH [125].
Recent investigations have highlighted the presence of circulating miRNA gradients across the pulmonary and cardiac vasculature in PAH. In a pediatric cohort, plasma samples were prospectively collected during cardiac catheterization from the superior vena cava, pulmonary artery, and ascending aorta in 12 children with PAH and nine matched controls. Unbiased profiling of 754 miRNAs revealed distinct trans-right-ventricle and transpulmonary gradients: miR-193a-5p and miR-423-5p exhibited opposing patterns between PAH patients and controls, while miR-26b-5p and miR-331-3p demonstrated transpulmonary differences. Additional miRNAs, including miR-29a-3p, miR-26a-5p, miR-590-5p, and miR-200c-3p, were upregulated in PAH superior vena cava samples, correlating with prognostic hemodynamic parameters [122].
In an earlier study conducted on adult patients, circulating levels of muscle-specific miR-204 were evaluated at multiple sites along the pulmonary vasculature [126]. In WHO Group I PAH, miR-204 increased sequentially across the pulmonary circulation, while intracellular miR-204 in pulmonary artery smooth muscle cells was reduced, suggesting selective excretion by diseased cells. Peripheral blood mononuclear cells showed no significant changes, highlighting the tissue-specific nature of these alterations. These findings reinforce the concept that circulating miRNA gradients reflect underlying cellular pathobiology and differ between PH subtypes, offering potential as biomarkers and therapeutic targets.

4.2.5. Bicuspid Aortic Valve

BAV is the most common congenital heart defect found in adults. In some cases, it is concomitant to other CHDs, especially aortic ones [127,128]. Other hemodynamic properties characterize a standard aortic valve with three leaflets; therefore, BAV might lead to aortic root dilatation and increased shear stress on the aorta, leading to various complications [128].
There is still a limited amount of data concerning pediatric cohorts in the case of the role of miRNA in BAV. In one study, circulating miRNAs were proposed as early biomarkers of aortic remodeling. In children with BAV, serum miR-130a was significantly reduced in patients with aortic dilatation, inversely correlating with aortic diameter and implicating TGF-β signaling in disease progression. These data indicate that miR-130a can be used as a potential marker to differentiate patients into high and low risk [129].
More studies were performed in adult settings. Several circulating miRNAs have been linked to BAV-associated aortopathy. Aortic tissue profiling revealed upregulation of let-7e-5p and miR-196a-5p and downregulation of miR-17a-5p in BAV with dilatation, with correlations to HDL-C and valvular function, highlighting their biomarker potential [130]. Another study showed that miR-21, miR-133a, miR-143, and miR-145 are consistently altered in plasma and tissue, supporting their role as systemic indicators of proximal aortic wall remodeling [131]. Tissue-level investigations expanded on these findings. Whole-miRNome sequencing identified 12 dysregulated miRNAs in BAV-related thoracic aortic aneurysm, with downregulation of miR-424-3p and miR-3688-3p associated with Hippo, ErbB, TGF-β, and focal adhesion signaling [132]. Regional profiling of the ascending aorta showed differential expression of miR-128-3p, miR-150-5p, and miR-199b-5p between dilated and nondilated segments, linking miRNA regulation to wall shear stress and extracellular matrix remodeling [133]. Earlier array studies on stenotic vs. regurgitant aortic valves found reduced expression of miR-26a, miR-30b, and miR-195 in stenotic valves, with functional assays confirming roles in calcification pathways, suggesting a contribution to valvular mineralization [134]. Clinical significance, functions and changes of expression pattern in various cardiac diseases of some key miRNAs are summarized in Table 2.

4.3. Heart Failure

In pediatric heart failure, circulating and myocardial miRNA signatures display distinct profiles from adult heart failure. Stauffer et al. demonstrated that the miRNA profile in the hearts of children with dilated cardiomyopathy differs significantly from that of adult HF patients. They identified 17 unique miRNAs that exhibit altered or even opposite regulation in children compared to the adult population. Notably, certain miRNAs correlating with clinical responses to phosphodiesterase inhibition and Smad4 normalization, highlighting the necessity for age-specific molecular stratification. Oxidative stress–responsive miRNAs orchestrate mitochondrial integrity, antioxidant defenses, iron metabolism, ferroptosis, and survival signaling, functioning as both biomarkers and active modulators of heart failure progression. In children with advanced heart failure undergoing ventricular assist device implantation, longitudinal analyses revealed dynamic regulation of six circulating miRNAs linked to hemostatic pathways, including the downregulation of miR-409-3p, a negative regulator of coagulation factors 7 and 2, potentially reflecting a universal pro-thrombotic phenotype post-implantation [135,136,137,138].

4.4. Arrythmias in Children

4.4.1. Supraventricular and Ventricular Arrhythmias

miR-1 and the miR-133 family stand out as regulators of cardiac excitability and conduction [139,140]. Two recent studies explored their diagnostic potential in pediatric populations presenting with either ventricular or supraventricular arrhythmias. In both studies, molecular analysis was carried out on serum samples obtained from children and adolescents diagnosed with arrhythmias, compared against healthy controls. Expression profiling focused on miR-1, miR-133a, and miR-133b [141,142]. The findings were consistent across both investigations.
Elevated levels of miR-1 and miR-133a were observed in the SVA subgroup compared to controls, highlighting a potential link between these miRNAs and supraventricular rhythm disturbances. Notably, miR-133a expression was also significantly higher in supraventricular compared with ventricular arrhythmias, suggesting subtype-specific regulation. In contrast, miR-133b was uniformly downregulated in both arrhythmic groups relative to healthy individuals, pointing to a broader role in arrhythmogenesis regardless of arrhythmia type [141,142]. miR-1 and miR-133a demonstrated moderate to strong discriminative ability in distinguishing supraventricular arrhythmia patients from arrhythmia-free controls. At the same time, miR-133b showed utility in differentiating both supraventricular and ventricular arrhythmia patients from healthy cohorts. These observations support the notion that distinct miRNA signatures may underlie specific arrhythmic mechanisms in pediatric populations [141].
Those results indicate that serum miR-1, miR-133a, and miR-133b could emerge as non-invasive biomarkers for pediatric arrhythmias. While miR-1 and miR-133a appear more closely associated with supraventricular arrhythmias, the consistent reduction in miR-133b across both arrhythmia types suggests its potential as a general marker of arrhythmic pathology. As both mentioned studies had limited sample sizes, further large-scale, multi-center studies are needed to validate these findings and to assess whether combining these miRNAs into a diagnostic panel could improve clinical accuracy.

4.4.2. Long QT Syndrome

Long QT syndrome is a group of genetic disorders caused by mutations in ion channels (sodium, potassium, calcium), which could lead to lethal arrhythmias [143]. The key to its treatment was early diagnosis and prevention of those fatal rhythm disturbances either by pharmacotherapy or implantation of implantable cardioverter defibrillators in patients who were resuscitated before [143]. Studies conducted on animal models suggested the role of miR-1 and miR-133 in the development of QT-prolongation [144,145]. These miRNAs are involved in the regulation of ion channels crucial for ventricular repolarization. MiRNA-1 inhibits the expression of the GJA1 and KCNJ2 genes encoding Cx43, responsible for intracellular conductance in ventricles, and the K+ channel subunit Kir2.1, which maintains the resting membrane potential [2]. In turn, in miR-1-2 knockout mice, a decrease in KCND2 expression was observed due to the lack of negative regulation of its repressor protein Irx5. The authors concluded that loss of Irx5 may lead to disrupted ventricular repolarization, given that the KCND2 product, K+ channel subunit (Kv4.2), is responsible for the transient outward K+ current [2]. Rau et al. proved that disturbances in the biogenesis of miR-1 lead to increased expression of CACNA1C. In the case of miR-133a, it was shown to inhibit KCNQ1, a miRNA that encodes Kv7.1 [146]. It is responsible for the repolarization phase, and loss of its function leads to the development of LQTS [147]. Both miR-1 and miR-133 are responsible for increased phosphorylation of the ryanodine receptor, and their dysregulation leads to arrhythmogenesis [77]. Hendley and colleagues decided to investigate the impact of specific sequence variants of those miRNAs (miR-1 and miR-133 families) on mature miRNA products and their relation to long QT syndrome development [148]. None of the found variants affected the mature miRNA products in the investigated cohort of patients’ samples; therefore, the authors suggested another mechanism of QT prolongation in this cohort.

4.4.3. miRNA as a Prognostic Tool of Arrhythmia Recurrence After Ablation

Multiple studies were conducted on the topic of miRNA changes after ablation of atrial fibrillation [149]. The ablation seems to restore certain miRNA levels to its base values; therefore, it was investigated whether they could serve as potential prognostic biomarkers of AF recurrence [149,150]. This topic was not investigated in case of pediatric cohorts nor any of the arrhythmias mentioned in this paragraph, but based on alterations of miRNA profiles in patients suffering from arrhythmias, some changes might be present post-ablation, and this topic and its clinical significance should be investigated in future studies.

4.5. Cardiomyopathies

miRNAs have emerged as critical post-transcriptional regulators in pediatric dilated cardiomyopathy, with roles spanning disease pathogenesis, biomarker discovery, and therapeutic innovation. Transcriptomic profiling of myocardial tissue from affected children revealed extensive age-dependent dysregulation of miRNAs, with over 390 species altered relative to non-failing controls, influencing pathways in stem cell differentiation, sarcomeric function, and intracellular signaling. Comparative analyses of paired myocardial and serum samples demonstrated that a subset of dysregulated miRNAs is consistently altered in both compartments, implicating circulating miRNAs as potential surrogates for myocardial expression changes and highlighting their involvement in processes such as inflammation, mitochondrial function, and metabolism. Prognostically, distinct circulating miRNA signatures, including upregulation of hsa-miR-155 and hsa-miR-636 and downregulation of hsa-miR-646 and hsa-miR-639, discriminate between children requiring transplantation or experiencing mortality and those with functional recovery, achieving high predictive accuracy. Therapeutically, cardiosphere-derived cells and their exosomes have shown promise in preclinical cardiomyopathy models, where exosomes enriched in cardioprotective miRNAs, notably miR-146a-5p, reduced myocardial fibrosis and improved ventricular function, findings preliminarily supported in early-phase pediatric trials. Independent profiling of plasma miRNAs has further identified distinct dysregulation patterns—including increased miR-518f and miR-454 and reduced miR-618, miR-875-3p, miR-205, miR-194, miR-302a, miR-147, and miR-544—which may represent additional diagnostic candidates for pediatric cardiomyopathy. Finally, circulating miRNA panels such as miR-142-5p, miR-143-3p, miR-27b-3p, and miR-126-3p have demonstrated diagnostic potential for childhood cardiomyopathy, with miR-126-3p and let-7g levels correlating inversely with left ventricular ejection fraction and distinguishing heart failure from non-heart failure phenotypes. Collectively, these data position miRNAs at the intersection of mechanistic insight, diagnostic precision, and emerging biologic therapy in pediatric cardiomyopathy [82,151,152,153,154].

4.6. Carditis

Carditis, spanning viral and rheumatic etiologies, reflects a complex convergence of myocardial injury and inflammation, with miRNAs emerging as pivotal regulatory nodes. In pediatric viral myocarditis, circulating miR-133b is markedly downregulated and inversely correlates with myocardial damage, mitigating Coxsackievirus B3–induced cardiomyocyte injury by suppressing proliferation and restraining TNF-α and IL-6 secretion via direct targeting of Rab27B. Additional miRNAs, including miR-381, miR-217, and miR-543, govern cardiomyocyte apoptosis and inflammatory signaling through modulation of COX-2 and SIRT1, whereas cardiac-associated miR-208a and miR-21 acutely mirror active injury, and subacute miR-208b predicts subsequent left ventricular functional recovery. In pediatric rheumatic carditis, miR-16-5p, miR-223-3p, and miR-92a-3p are significantly downregulated, highlighting their potential as disease-specific biomarkers. Together, these data position miRNAs as central orchestrators of inflammatory and injury pathways in carditis, offering novel opportunities for precision diagnostics and targeted therapeutics [13,14,15,16,17].

5. Challenges and Future Perspectives

5.1. Challenges in Therapeutic Applications

One of the fundamental challenges in utilizing miRNAs therapeutically is their multi-targeting nature. A single gene can be regulated by multiple miRNAs, either directly through repression of its mRNA or through the repression of relevant transcription factors. Simultaneously, a single miRNA often influences multiple genes [155]. This complexity significantly complicates the prediction of therapeutic outcomes. An example is the attempt to use miR-34a in the treatment of certain cancers. The use of a miR-34a analog resulted in serious adverse effects related to the immune response, likely resulting from the activation of two immune-related pathways [156].
Another major obstacle is the delivery of miRNAs. They are highly susceptible to degradation by nucleases, have relatively low efficiency in targeted delivery to specific tissues, and the carriers used can provoke immunogenic responses [157]. Due to the presence of specific nucleases in serum, such as serum RNase A-type nuclease, the biological half-life in blood and cells differs significantly [158]. This problem can be overcome by chemical modifications of nucleotides, such as methylation or fluorination of the 2′ ribose [159].
To enhance delivery efficiency and protect miRNA from degradation, many specific carriers have been developed. Among them, lipid-based delivery systems are widely used [160]. However, their limited specificity makes targeted delivery to cardiac cells particularly challenging. Functionalization of liposomes has shown promising results. Attaching a specific peptide to their surface that binds to cardiomyocytes, particularly in areas of ischemia and remodeling, enabled targeted delivery of the miR-185-5p inhibitor. As a result, a significant improvement in EF and a reduction in fibrosis were achieved [159]. Another significant limitation is the low solubility and short half-life of liposomes. For this reason, the use of polymeric delivery systems such as polyethyleneimines (PEIs) has been explored. Due to their positive electrostatic charge, they strongly bind negatively charged miRNA molecules [161]. Due to their high cytotoxicity, clinical applications usually require combination with other polymers such as poly L-Lysine (PLL) or polyethylene glycol (PEG) [162,163]. Ease of aggregation and uncontrolled release of the transported substances are also problematic [161]. The use of extracellular vesicles (EVs) also offers hope for increased treatment efficacy. MiRNA-21 transported in this manner led to significant improvement in cardiac function in a preclinical MI animal model [164]. Nonetheless, the efficiency of miRNA loading into EVs is often inconsistent and low, and EV subpopulations exhibit distinct physicochemical and biological properties, which impacts the reproducibility of results and complicates dose control. Additionally, off-target uptake by phagocytic cells remains a significant problem [165]. The use of viral vectors, such as lentiviruses, adenoviruses, and adeno-associated viruses, is also popular. Unlike EVs, their significant advantage is high infection efficiency and greater specific tissue selectivity [166]. Injection of AAV9-anti-miR-199a reduced hypertrophy and increased ejection fraction in mice treated with isoproterenol [167]. Unfortunately, viral vectors exhibit relatively high immunogenicity, which may lead to damage to transfected cells and reduce the effectiveness of therapy due to binding to neutralizing antibodies [166]. In a phase 1/2 clinical trial in patients with HF, cases of AAV vector therapy failure due to the presence of the above-mentioned antibodies were observed [168].
Additionally, ultrasound-targeted microbubble cavitation (UTMC) is being explored. This technique involves the intravenous administration of lipid-protein vesicles filled with gas and a therapeutic and is followed by exposing the target tissue to ultrasound, which induces cavitation and cargo release. AntimiR-23a administered this way reduced cardiac hypertrophy in mice [169]. Local delivery of hydrogels containing polymer nanoparticles with miRNA has also been attempted. In a study of rats with myocardial infarction, administration of miR-199a-3p by this method resulted in an increase in ejection fraction and a reduction in scar size [170]. Unfortunately, the carriers themselves can also be toxic. Cationic liposomes, through electrostatic interactions with negatively charged membranes, destabilize them. Modifications that alter their surface charge, such as pegylation, reduce their cytotoxicity [171].
MiRNAs may be promising biomarkers for early disease detection. Currently, the main problem is the lack of standardization in analytical and preanalytical procedures. Measurements may not be reproducible due to low miRNA stability. Comparing studies becomes difficult due to the use of different protocols—miRNA levels are influenced by factors such as the type of anticoagulant, sample storage time, temperature, and the specific isolation method used [90].

5.2. Future Directions and Emerging Opportunities

Combining miRNA with other therapies can yield synergistic effects. Transduction of transplanted cardiac progenitor cells (CPCs) with miR-21, miR-24, and miR-221 after MI in mice increased their survival. Furthermore, improved left ventricular fractional shortening was observed compared to the group receiving unmodified CPCs. This effect is likely due to inhibition of Bim gene expression, a key activator of apoptosis.
The rapid development of artificial intelligence observed in recent years may contribute to significant progress in miRNA research. Neural networks can help predict miRNA-mRNA interactions, which contributes to improved sensitivity for noncanonical binding events [172]. A 2021 study using distinct machine learning approaches identified two circulating miRNAs that can aid in the diagnosis of pulmonary hypertension [173]. Thanks to increasingly advanced algorithms, a personalized multi-omics approach may become possible, where data based on miRNA levels, microbiome composition, and levels of specific metabolites will enable more precise diagnoses and prognoses [174].
Early attempts to detect miRNAs relied primarily on techniques such as PCR, which required the use of material obtained from multiple cells for a single measurement [175]. This made it difficult to determine cellular heterogeneity. The advent of single-cell miRNA sequencing (sc-miRNA-seq), which can provide information on miRNA populations in individual cells, was a breakthrough [176]. Importantly, integration with spatial transcriptomics allows not only the identification of biological processes in tissue but also the precise localization of these processes in specific tissue regions, enabling the creation of high-resolution maps of miRNA activity [177]. Currently, there is a lack of studies using AI and modern methods such as sc-miRNA-seq in pediatric cardiology. Further research is needed to implement these approaches in this field. Perhaps this approach will soon enable better assessment of cardiac tissue heterogeneity during development, mapping miRNA activity in regions affected by congenital defects, and better identifying potential biomarkers.

6. Conclusions

MiRNAs constitute a key regulatory link in heart development and proper function. They influence key cellular processes such as proliferation, differentiation, and apoptosis. A thorough understanding of the molecular mechanisms underlying these phenomena offers hope for the development of new diagnostic and therapeutic methods. In recent years, significant progress has been made in research on the use of miRNAs in the diagnosis and treatment of various conditions, such as congenital heart defects, arrhythmias, carditis, and heart failure in children. Unfortunately, research focused on their practical application in clinical medicine, including monitoring the course of cardiovascular disease, assessing therapeutic efficacy, and identifying new therapeutic targets, is still lacking.

Author Contributions

Conceptualization A.K., M.P., J.I.K., M.K. and J.B.; Validation, J.B. and M.K.; investigation, A.K., M.P. and J.I.K.; writing—original draft preparation, A.K., M.P. and J.I.K.; writing—review and editing, J.B. and M.K.; visualization, J.I.K.; supervision, J.B. and M.K.; project administration, J.B.; funding acquisition, J.I.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing does not apply to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ASDatrial septal defect
AgoArgonaute proteins
BAVbicuspid aortic valve
BMPbone morphogenic proteins
CPCscardiac progenitor cells
cTNTcardiac Troponin T
CoAcoarctation of the aorta
CHDcongenital heart disease
Cx43connexin 43
FGFfibroblast growth factor
hESCs-CMshuman embryonic Stem Cells–derived Cardiomyocytes
HLHShypoplastic left heart syndrome
miRNAmicroRNA
mRNAmessenger RNA
PDApatent ductus arteriosus
PABPCpoly(A)-binding protein, cytoplasmic
PEGpolyethylene glycol
PEIspolyethyleneimines
PLLpoly L-Lysine
Pol IIRNA polymerase II
RVright ventricle
RVOTright ventricular outflow tract
RISCRNA-induced silencing complex
sc-miRNA-seqsingle-cell microRNA sequencing
TDMDtarget-directed microRNA degradation
TAtricuspid atresia
TGAtransposition of the great arteries
TOFtetralogy of fallot
TET2Tet meyhylcytosine dioxygense 2
VSDventricular septal defect

References

  1. Cordes, K.R.; Srivastava, D.; Ivey, K.N. MicroRNAs in Cardiac Development. Pediatr. Cardiol. 2010, 31, 349–356. [Google Scholar] [CrossRef]
  2. Zhao, Y.; Ransom, J.F.; Li, A.; Vedantham, V.; von Drehle, M.; Muth, A.N.; Tsuchihashi, T.; McManus, M.T.; Schwartz, R.J.; Srivastava, D. Dysregulation of Cardiogenesis, Cardiac Conduction, and Cell Cycle in Mice Lacking MiRNA-1-2. Cell 2007, 129, 303–317. [Google Scholar] [CrossRef] [PubMed]
  3. Blue, G.M.; Kirk, E.P.; Sholler, G.F.; Harvey, R.P.; Winlaw, D.S. Congenital Heart Disease: Current Knowledge about Causes and Inheritance. Med. J. Aust. 2012, 197, 155–159. [Google Scholar] [CrossRef]
  4. Lipshultz, S.E.; Law, Y.M.; Asante-Korang, A.; Austin, E.D.; Dipchand, A.I.; Everitt, M.D.; Hsu, D.T.; Lin, K.Y.; Price, J.F.; Wilkinson, J.D.; et al. Cardiomyopathy in Children: Classification and Diagnosis: A Scientific Statement from the American Heart Association. Circulation 2019, 140, E9–E68. [Google Scholar] [CrossRef]
  5. Law, Y.M.; Lal, A.K.; Chen, S.; Čiháková, D.; Cooper, L.T.; Deshpande, S.; Godown, J.; Grosse-Wortmann, L.; Robinson, J.D.; Towbin, J.A. Diagnosis and Management of Myocarditis in Children. Circulation 2021, 144, E123–E135. [Google Scholar] [CrossRef] [PubMed]
  6. Abu-Halima, M.; Meese, E.; Keller, A.; Abdul-Khaliq, H.; Rädle-Hurst, T. Analysis of Circulating MicroRNAs in Patients with Repaired Tetralogy of Fallot with and without Heart Failure. J. Transl. Med. 2017, 15, 156. [Google Scholar] [CrossRef]
  7. Abu-Halima, M.; Meese, E.; Saleh, M.A.; Keller, A.; Abdul-Khaliq, H.; Raedle-Hurst, T. Micro-RNA 150-5p Predicts Overt Heart Failure in Patients with Univentricular Hearts. PLoS ONE 2019, 14, e0223606. [Google Scholar] [CrossRef]
  8. Ramachandran, S.; Lowenthal, A.; Ritner, C.; Lowenthal, S.; Bernstein, H.S. Plasma Microvesicle Analysis Identifies MicroRNA 129-5p as a Biomarker of Heart Failure in Univentricular Heart Disease. PLoS ONE 2017, 12, e0183624. [Google Scholar] [CrossRef]
  9. Sucharov, C.C.; Sucharov, J.; Karimpour-Fard, A.; Nunley, K.; Stauffer, B.L.; Miyamoto, S.D. Micro-RNA Expression in Hypoplastic Left Heart Syndrome. J. Card. Fail. 2015, 21, 83–88. [Google Scholar] [CrossRef]
  10. Li, J.; Cao, Y.; Ma, X.; Wang, H.; Zhang, J.; Luo, X.; Chen, W.; Wu, Y.; Meng, Y.; Zhang, J.; et al. Roles of MiR-1-1 and MiR-181c in Ventricular Septal Defects. Int. J. Cardiol. 2013, 168, 1441–1446. [Google Scholar] [CrossRef] [PubMed]
  11. Song, Y.; Higgins, H.; Guo, J.; Harrison, K.; Schultz, E.N.; Hales, B.J.; Moses, E.K.; Goldblatt, J.; Pachter, N.; Zhang, G. Clinical Significance of Circulating MicroRNAs as Markers in Detecting and Predicting Congenital Heart Defects in Children. J. Transl. Med. 2018, 16, 42. [Google Scholar] [CrossRef]
  12. Latronico, M.V.G.; Catalucci, D.; Condorelli, G. MicroRNA and Cardiac Pathologies. Physiol. Genom. 2008, 34, 239–242. [Google Scholar] [CrossRef]
  13. Goldberg, L.; Tirosh-Wagner, T.; Vardi, A.; Abbas, H.; Pillar, N.; Shomron, N.; Nevo-Caspi, Y.; Paret, G. Circulating MicroRNAs: A Potential Biomarker for Cardiac Damage, Inflammatory Response, and Left Ventricular Function Recovery in Pediatric Viral Myocarditis. J. Cardiovasc. Transl. Res. 2018, 11, 319–328. [Google Scholar] [CrossRef] [PubMed]
  14. Zhang, Y.; Sun, L.; Sun, H.; Yu, Z.; Liu, X.; Luo, X.; Li, C.; Sun, D.; Li, T. MicroRNA-381 Protects Myocardial Cell Function in Children and Mice with Viral Myocarditis via Targeting Cyclooxygenase-2 Expression. Exp. Ther. Med. 2018, 15, 5510–5516. [Google Scholar] [CrossRef] [PubMed]
  15. Xia, K.; Zhang, Y.; Sun, D. MiR-217 and MiR-543 Downregulation Mitigates Inflammatory Response and Myocardial Injury in Children with Viral Myocarditis by Regulating the SIRT1/AMPK/NF-κB Signaling Pathway. Int. J. Mol. Med. 2019, 45, 634–646. [Google Scholar] [CrossRef] [PubMed]
  16. He, F.; Liu, Z.; Feng, M.; Xiao, Z.; Yi, X.; Wu, J.; Liu, Z.; Wang, G.; Li, L.; Yao, H. The LncRNA MEG3/MiRNA-21/P38MAPK Axis Inhibits Coxsackievirus 3 Replication in Acute Viral Myocarditis. Virus Res. 2024, 339, 199250. [Google Scholar] [CrossRef]
  17. Zhang, X.; Yang, R.; Ma, M.; Chen, S.; Guo, K.; Zhang, L.; You, Y.; Jia, H.; Han, B. Altered Plasma Exosome MiRNAs and Novel Potential Biomarkers in Pediatric Fulminant Myocarditis. Genomics 2023, 115, 110622. [Google Scholar] [CrossRef]
  18. Lee, R.C.; Feinbaum, R.L.; Ambros, V. The C. elegans Heterochronic Gene Lin-4 Encodes Small RNAs with Antisense Complementarity to Lin-14. Cell 1993, 75, 843–854. [Google Scholar] [CrossRef]
  19. Reinhart, B.J.; Slack, F.J.; Basson, M.; Pasquienelll, A.E.; Bettlnger, J.C.; Rougvle, A.E.; Horvitz, H.R.; Ruvkun, G. The 21-Nucleotide Let-7 RNA Regulates Developmental Timing in Caenorhabditis Elegans. Nature 2000, 403, 901–906. [Google Scholar] [CrossRef]
  20. Loganathan, T.; Doss C, G.P. Non-Coding RNAs in Human Health and Disease: Potential Function as Biomarkers and Therapeutic Targets. Funct. Integr. Genom. 2023, 23, 33. [Google Scholar] [CrossRef]
  21. Bartel, D.P. Metazoan MicroRNAs. Cell 2018, 173, 20–51. [Google Scholar] [CrossRef]
  22. Lee, Y.; Kim, M.; Han, J.; Yeom, K.H.; Lee, S.; Baek, S.H.; Kim, V.N. MicroRNA Genes Are Transcribed by RNA Polymerase II. EMBO J. 2004, 23, 4051–4060. [Google Scholar] [CrossRef]
  23. Lee, Y.; Ahn, C.; Han, J.; Choi, H.; Kim, J.; Yim, J.; Lee, J.; Provost, P.; Rådmark, O.; Kim, S.; et al. The Nuclear RNase III Drosha Initiates MicroRNA Processing. Nature 2003, 425, 415–419. [Google Scholar] [CrossRef]
  24. Lund, E.; Güttinger, S.; Calado, A.; Dahlberg, J.E.; Kutay, U. Nuclear Export of MicroRNA Precursors. Science 2004, 303, 95–98. [Google Scholar] [CrossRef]
  25. Zhang, H.; Kolb, F.A.; Jaskiewicz, L.; Westhof, E.; Filipowicz, W. Single Processing Center Models for Human Dicer and Bacterial RNase III. Cell 2004, 118, 57–68. [Google Scholar] [CrossRef]
  26. Iwasaki, S.; Kobayashi, M.; Yoda, M.; Sakaguchi, Y.; Katsuma, S.; Suzuki, T.; Tomari, Y. Hsc70/Hsp90 Chaperone Machinery Mediates ATP-Dependent RISC Loading of Small RNA Duplexes. Mol. Cell 2010, 39, 292–299. [Google Scholar] [CrossRef]
  27. Frank, F.; Sonenberg, N.; Nagar, B. Structural Basis for 5′-Nucleotide Base-Specific Recognition of Guide RNA by Human AGO2. Nature 2010, 465, 818–822. [Google Scholar] [CrossRef] [PubMed]
  28. Iwakawa, H.; Tomari, Y. Life of RISC: Formation, Action, and Degradation of RNA-Induced Silencing Complex. Mol. Cell 2022, 82, 30–43. [Google Scholar] [CrossRef] [PubMed]
  29. Ruby, J.G.; Jan, C.H.; Bartel, D.P. Intronic MicroRNA Precursors That Bypass Drosha Processing. Nature 2007, 448, 83–86. [Google Scholar] [CrossRef] [PubMed]
  30. Cheloufi, S.; Dos Santos, C.O.; Chong, M.M.W.; Hannon, G.J. A Dicer-Independent MiRNA Biogenesis Pathway That Requires Ago Catalysis. Nature 2010, 465, 584–589. [Google Scholar] [CrossRef]
  31. Ozsolak, F.; Poling, L.L.; Wang, Z.; Liu, H.; Liu, X.S.; Roeder, R.G.; Zhang, X.; Song, J.S.; Fisher, D.E. Chromatin Structure Analyses Identify MiRNA Promoters. Genes Dev. 2008, 22, 3172–3183. [Google Scholar] [CrossRef]
  32. Ha, M.; Kim, V.N. Regulation of MicroRNA Biogenesis. Nat. Rev. Mol. Cell Biol. 2014, 15, 509–524. [Google Scholar] [CrossRef] [PubMed]
  33. Heo, I.; Joo, C.; Cho, J.; Ha, M.; Han, J.; Kim, V.N. Lin28 Mediates the Terminal Uridylation of Let-7 Precursor MicroRNA. Mol. Cell 2008, 32, 276–284. [Google Scholar] [CrossRef] [PubMed]
  34. Heo, I.; Ha, M.; Lim, J.; Yoon, M.-J.; Park, J.-E.; Kwon, S.C.; Chang, H.; Kim, V.N. Mono-Uridylation of Pre-MicroRNA as a Key Step in the Biogenesis of Group II Let-7 MicroRNAs. Cell 2012, 151, 521–532. [Google Scholar] [CrossRef]
  35. Yang, W.; Chendrimada, T.P.; Wang, Q.; Higuchi, M.; Seeburg, P.H.; Shiekhattar, R.; Nishikura, K. Modulation of MicroRNA Processing and Expression through RNA Editing by ADAR Deaminases. Nat. Struct. Mol. Biol. 2006, 13, 13–21. [Google Scholar] [CrossRef]
  36. Michlewski, G.; Cáceres, J.F. Post-Transcriptional Control of MiRNA Biogenesis. RNA 2019, 25, 1–16. [Google Scholar] [CrossRef] [PubMed]
  37. Vieux, K.-F.; Prothro, K.P.; Kelley, L.H.; Palmer, C.; Maine, E.M.; Veksler-Lublinsky, I.; McJunkin, K. Screening by Deep Sequencing Reveals Mediators of MicroRNA Tailing in C. elegans. Nucleic Acids Res. 2021, 49, 11167–11180. [Google Scholar] [CrossRef]
  38. Bail, S.; Swerdel, M.; Liu, H.; Jiao, X.; Goff, L.A.; Hart, R.P.; Kiledjian, M. Differential Regulation of MicroRNA Stability. RNA 2010, 16, 1032–1039. [Google Scholar] [CrossRef]
  39. Krol, J.; Busskamp, V.; Markiewicz, I.; Stadler, M.B.; Ribi, S.; Richter, J.; Duebel, J.; Bicker, S.; Fehling, H.J.; Schübeler, D.; et al. Characterizing Light-Regulated Retinal MicroRNAs Reveals Rapid Turnover as a Common Property of Neuronal MicroRNAs. Cell 2010, 141, 618–631. [Google Scholar] [CrossRef]
  40. de la Mata, M.; Gaidatzis, D.; Vitanescu, M.; Stadler, M.B.; Wentzel, C.; Scheiffele, P.; Filipowicz, W.; Großhans, H. Potent Degradation of Neuronal MiRNAs Induced by Highly Complementary Targets. EMBO Rep. 2015, 16, 500–511. [Google Scholar] [CrossRef]
  41. Shin, C.; Nam, J.-W.; Farh, K.K.-H.; Chiang, H.R.; Shkumatava, A.; Bartel, D.P. Expanding the MicroRNA Targeting Code: Functional Sites with Centered Pairing. Mol. Cell 2010, 38, 789–802. [Google Scholar] [CrossRef] [PubMed]
  42. Wee, L.M.; Flores-Jasso, C.F.; Salomon, W.E.; Zamore, P.D. Argonaute Divides Its RNA Guide into Domains with Distinct Functions and RNA-Binding Properties. Cell 2012, 151, 1055–1067. [Google Scholar] [CrossRef] [PubMed]
  43. Nakanishi, K. When Argonaute Takes out the Ribonuclease Sword. J. Biol. Chem. 2024, 300, 105499. [Google Scholar] [CrossRef] [PubMed]
  44. Rehwinkel, J.; Behm-Ansmant, I.; Gatfield, D.; Izaurralde, E. A Crucial Role for GW182 and the DCP1:DCP2 Decapping Complex in MiRNA-Mediated Gene Silencing. RNA 2005, 11, 1640–1647. [Google Scholar] [CrossRef]
  45. Jonas, S.; Izaurralde, E. Towards a Molecular Understanding of MicroRNA-Mediated Gene Silencing. Nat. Rev. Genet. 2015, 16, 421–433. [Google Scholar] [CrossRef]
  46. Chen, C.A.; Shyu, A. Mechanisms of Deadenylation-dependent Decay. WIREs RNA 2011, 2, 167–183. [Google Scholar] [CrossRef]
  47. Kamenska, A.; Lu, W.-T.; Kubacka, D.; Broomhead, H.; Minshall, N.; Bushell, M.; Standart, N. Human 4E-T Represses Translation of Bound MRNAs and Enhances MicroRNA-Mediated Silencing. Nucleic Acids Res. 2014, 42, 3298–3313. [Google Scholar] [CrossRef]
  48. Lewis, B.P.; Burge, C.B.; Bartel, D.P. Conserved Seed Pairing, Often Flanked by Adenosines, Indicates That Thousands of Human Genes Are MicroRNA Targets. Cell 2005, 120, 15–20. [Google Scholar] [CrossRef]
  49. Alberti, C.; Cochella, L. A Framework for Understanding the Roles of MiRNAs in Animal Development. Development 2017, 144, 2548–2559. [Google Scholar] [CrossRef]
  50. Ouyang, Z.; Wei, K. MiRNA in Cardiac Development and Regeneration. Cell Regen. 2021, 10, 14. [Google Scholar] [CrossRef]
  51. Saxena, A.; Tabin, C.J. MiRNA-Processing Enzyme Dicer Is Necessary for Cardiac Outflow Tract Alignment and Chamber Septation. Proc. Natl. Acad. Sci. USA 2010, 107, 87–91. [Google Scholar] [CrossRef]
  52. da Costa Martins, P.A.; Bourajjaj, M.; Gladka, M.; Kortland, M.; van Oort, R.J.; Pinto, Y.M.; Molkentin, J.D.; De Windt, L.J. Conditional Dicer Gene Deletion in the Postnatal Myocardium Provokes Spontaneous Cardiac Remodeling. Circulation 2008, 118, 1567–1576. [Google Scholar] [CrossRef]
  53. Lagos-Quintana, M.; Rauhut, R.; Yalcin, A.; Meyer, J.; Lendeckel, W.; Tuschl, T. Identification of Tissue-Specific MicroRNAs from Mouse. Curr. Biol. 2002, 12, 735–739. [Google Scholar] [CrossRef]
  54. Sylva, M.; van den Hoff, M.J.B.; Moorman, A.F.M. Development of the Human Heart. Am. J. Med. Genet. A 2014, 164, 1347–1371. [Google Scholar] [CrossRef]
  55. Pisano, F.; Altomare, C.; Cervio, E.; Barile, L.; Rocchetti, M.; Ciuffreda, M.C.; Malpasso, G.; Copes, F.; Mura, M.; Danieli, P.; et al. Combination of MiRNA499 and MiRNA133 Exerts a Synergic Effect on Cardiac Differentiation. Stem Cells 2015, 33, 1187–1199. [Google Scholar] [CrossRef]
  56. van Rooij, E.; Quiat, D.; Johnson, B.A.; Sutherland, L.B.; Qi, X.; Richardson, J.A.; Kelm, R.J.; Olson, E.N. A Family of MicroRNAs Encoded by Myosin Genes Governs Myosin Expression and Muscle Performance. Dev. Cell 2009, 17, 662–673. [Google Scholar] [CrossRef]
  57. Li, K.-X.; Li, J.-R.; Zuo, S.-J.; Li, X.; Chen, X.-T.; Xiao, P.-Y.; Li, H.-T.; Sun, L.; Qian, T.; Zhang, H.-M.; et al. Identification of MiR-20b-5p as an Inhibitory Regulator in Cardiac Differentiation via TET2 and DNA Hydroxymethylation. Clin. Epigenetics 2024, 16, 42. [Google Scholar] [CrossRef] [PubMed]
  58. Hoelscher, S.C.; Stich, T.; Diehm, A.; Lahm, H.; Dreßen, M.; Zhang, Z.; Neb, I.; Aherrahrou, Z.; Erdmann, J.; Schunkert, H.; et al. MiR-128a Acts as a Regulator in Cardiac Development by Modulating Differentiation of Cardiac Progenitor Cell Populations. Int. J. Mol. Sci. 2020, 21, 1158. [Google Scholar] [CrossRef] [PubMed]
  59. Porrello, E.R.; Johnson, B.A.; Aurora, A.B.; Simpson, E.; Nam, Y.-J.; Matkovich, S.J.; Dorn, G.W.; van Rooij, E.; Olson, E.N. MiR-15 Family Regulates Postnatal Mitotic Arrest of Cardiomyocytes. Circ. Res. 2011, 109, 670–679. [Google Scholar] [CrossRef] [PubMed]
  60. Gao, Z.; Zhu, X.; Dou, Y. The MiR-302/367 Cluster: A Comprehensive Update on Its Evolution and Functions. Open Biol. 2015, 5, 150138. [Google Scholar] [CrossRef]
  61. Chen, J.; Huang, Z.-P.; Seok, H.Y.; Ding, J.; Kataoka, M.; Zhang, Z.; Hu, X.; Wang, G.; Lin, Z.; Wang, S.; et al. Mir-17–92 Cluster Is Required for and Sufficient to Induce Cardiomyocyte Proliferation in Postnatal and Adult Hearts. Circ. Res. 2013, 112, 1557–1566. [Google Scholar] [CrossRef]
  62. Clark, A.L.; Naya, F.J. MicroRNAs in the Myocyte Enhancer Factor 2 (MEF2)-Regulated Gtl2-Dio3 Noncoding RNA Locus Promote Cardiomyocyte Proliferation by Targeting the Transcriptional Coactivator Cited2. J. Biol. Chem. 2015, 290, 23162–23172. [Google Scholar] [CrossRef]
  63. Raso, A.; Dirkx, E.; Sampaio-Pinto, V.; el Azzouzi, H.; Cubero, R.J.; Sorensen, D.W.; Ottaviani, L.; Olieslagers, S.; Huibers, M.M.; de Weger, R.; et al. A MicroRNA Program Regulates the Balance between Cardiomyocyte Hyperplasia and Hypertrophy and Stimulates Cardiac Regeneration. Nat. Commun. 2021, 12, 4808. [Google Scholar] [CrossRef]
  64. Renikunta, H.V.; Lazarow, K.; Gong, Y.; Shukla, P.C.; Nageswaran, V.; Giral, H.; Kratzer, A.; Opitz, L.; Engel, F.B.; Haghikia, A.; et al. Large-Scale MicroRNA Functional High-Throughput Screening Identifies MiR-515-3p and MiR-519e-3p as Inducers of Human Cardiomyocyte Proliferation. iScience 2023, 26, 106593. [Google Scholar] [CrossRef]
  65. Fish, J.E.; Santoro, M.M.; Morton, S.U.; Yu, S.; Yeh, R.-F.; Wythe, J.D.; Ivey, K.N.; Bruneau, B.G.; Stainier, D.Y.R.; Srivastava, D. MiR-126 Regulates Angiogenic Signaling and Vascular Integrity. Dev. Cell 2008, 15, 272–284. [Google Scholar] [CrossRef] [PubMed]
  66. Hu, S.; Huang, M.; Li, Z.; Jia, F.; Ghosh, Z.; Lijkwan, M.A.; Fasanaro, P.; Sun, N.; Wang, X.; Martelli, F.; et al. MicroRNA-210 as a Novel Therapy for Treatment of Ischemic Heart Disease. Circulation 2010, 122 (Suppl. S1), S124–S131. [Google Scholar] [CrossRef] [PubMed]
  67. Rogg, E.-M.; Abplanalp, W.T.; Bischof, C.; John, D.; Schulz, M.H.; Krishnan, J.; Fischer, A.; Poluzzi, C.; Schaefer, L.; Bonauer, A.; et al. Analysis of Cell Type-Specific Effects of MicroRNA-92a Provides Novel Insights Into Target Regulation and Mechanism of Action. Circulation 2018, 138, 2545–2558. [Google Scholar] [CrossRef] [PubMed]
  68. Gallant-Behm, C.L.; Piper, J.; Dickinson, B.A.; Dalby, C.M.; Pestano, L.A.; Jackson, A.L. A Synthetic MicroRNA-92a Inhibitor (MRG-110) Accelerates Angiogenesis and Wound Healing in Diabetic and Nondiabetic Wounds. Wound Repair. Regen. 2018, 26, 311–323. [Google Scholar] [CrossRef]
  69. Boettger, T.; Beetz, N.; Kostin, S.; Schneider, J.; Krüger, M.; Hein, L.; Braun, T. Acquisition of the Contractile Phenotype by Murine Arterial Smooth Muscle Cells Depends on the Mir143/145 Gene Cluster. J. Clin. Investig. 2009, 119, 2634–2647. [Google Scholar] [CrossRef]
  70. Mohan, R.A.; Boukens, B.J.; Christoffels, V.M. Developmental Origin of the Cardiac Conduction System: Insight from Lineage Tracing. Pediatr. Cardiol. 2018, 39, 1107–1114. [Google Scholar] [CrossRef]
  71. Wang, J.; Bai, Y.; Li, N.; Ye, W.; Zhang, M.; Greene, S.B.; Tao, Y.; Chen, Y.; Wehrens, X.H.T.; Martin, J.F. Pitx2-MicroRNA Pathway That Delimits Sinoatrial Node Development and Inhibits Predisposition to Atrial Fibrillation. Proc. Natl. Acad. Sci. USA 2014, 111, 9181–9186. [Google Scholar] [CrossRef] [PubMed]
  72. Petkova, M.; Atkinson, A.J.; Yanni, J.; Stuart, L.; Aminu, A.J.; Ivanova, A.D.; Pustovit, K.B.; Geragthy, C.; Feather, A.; Li, N.; et al. Identification of Key Small Non-Coding MicroRNAs Controlling Pacemaker Mechanisms in the Human Sinus Node. J. Am. Heart Assoc. 2020, 9, e016590. [Google Scholar] [CrossRef]
  73. Yanni, J.; D’Souza, A.; Wang, Y.; Li, N.; Hansen, B.J.; Zakharkin, S.O.; Smith, M.; Hayward, C.; Whitson, B.A.; Mohler, P.J.; et al. Silencing MiR-370-3p Rescues Funny Current and Sinus Node Function in Heart Failure. Sci. Rep. 2020, 10, 11279. [Google Scholar] [CrossRef]
  74. D’Souza, A.; Pearman, C.M.; Wang, Y.; Nakao, S.; Logantha, S.J.R.J.; Cox, C.; Bennett, H.; Zhang, Y.; Johnsen, A.B.; Linscheid, N.; et al. Targeting MiR-423-5p Reverses Exercise Training–Induced HCN4 Channel Remodeling and Sinus Bradycardia. Circ. Res. 2017, 121, 1058–1068. [Google Scholar] [CrossRef]
  75. Belevych, A.E.; Sansom, S.E.; Terentyeva, R.; Ho, H.-T.; Nishijima, Y.; Martin, M.M.; Jindal, H.K.; Rochira, J.A.; Kunitomo, Y.; Abdellatif, M.; et al. MicroRNA-1 and -133 Increase Arrhythmogenesis in Heart Failure by Dissociating Phosphatase Activity from RyR2 Complex. PLoS ONE 2011, 6, e28324. [Google Scholar] [CrossRef]
  76. Yang, B.; Lin, H.; Xiao, J.; Lu, Y.; Luo, X.; Li, B.; Zhang, Y.; Xu, C.; Bai, Y.; Wang, H.; et al. The Muscle-Specific MicroRNA MiR-1 Regulates Cardiac Arrhythmogenic Potential by Targeting GJA1 and KCNJ2. Nat. Med. 2007, 13, 486–491. [Google Scholar] [CrossRef] [PubMed]
  77. Callis, T.E.; Pandya, K.; Seok, H.Y.; Tang, R.-H.; Tatsuguchi, M.; Huang, Z.-P.; Chen, J.-F.; Deng, Z.; Gunn, B.; Shumate, J.; et al. MicroRNA-208a Is a Regulator of Cardiac Hypertrophy and Conduction in Mice. J. Clin. Investig. 2009, 119, 2772–2786. [Google Scholar] [CrossRef]
  78. Wei, Y.; Peng, S.; Wu, M.; Sachidanandam, R.; Tu, Z.; Zhang, S.; Falce, C.; Sobie, E.A.; Lebeche, D.; Zhao, Y. Multifaceted Roles of MiR-1s in Repressing the Fetal Gene Program in the Heart. Cell Res. 2014, 24, 278–292. [Google Scholar] [CrossRef]
  79. Lu, T.-Y.; Lin, B.; Li, Y.; Arora, A.; Han, L.; Cui, C.; Coronnello, C.; Sheng, Y.; Benos, P.V.; Yang, L. Overexpression of MicroRNA-1 Promotes Cardiomyocyte Commitment from Human Cardiovascular Progenitors via Suppressing WNT and FGF Signaling Pathways. J. Mol. Cell. Cardiol. 2013, 63, 146–154. [Google Scholar] [CrossRef]
  80. Li, X.; Bi, H.; Xie, S.; Cui, W. MiR-208b Regulates the Conversion of Skeletal Muscle Fiber Types by Inhibiting Mettl8 Expression. Front. Genet. 2022, 13, 820464. [Google Scholar] [CrossRef] [PubMed]
  81. Yang, Y.; Yang, H.; Lian, X.; Yang, S.; Shen, H.; Wu, S.; Wang, X.; Lyu, G. Circulating MicroRNA: Myocardium-Derived Prenatal Biomarker of Ventricular Septal Defects. Front. Genet. 2022, 13, 899034. [Google Scholar] [CrossRef]
  82. Miyamoto, S.D.; Karimpour-Fard, A.; Peterson, V.; Auerbach, S.R.; Stenmark, K.R.; Stauffer, B.L.; Sucharov, C.C. Circulating MicroRNA as a Biomarker for Recovery in Pediatric Dilated Cardiomyopathy. J. Heart Lung Transplant. 2015, 34, 724–733. [Google Scholar] [CrossRef] [PubMed]
  83. Chen, W.; Li, S. Circulating MicroRNA as a Novel Biomarker for Pulmonary Arterial Hypertension Due to Congenital Heart Disease. Pediatr. Cardiol. 2017, 38, 86–94. [Google Scholar] [CrossRef] [PubMed]
  84. Jin, Y.; Ai, L.; Chai, X.; Tang, P.; Zhang, W.; Yang, L.; Hu, Y.; Xu, Y.; Li, S. Maternal Circulating Exosomal MiRNAs as Non-Invasive Biomarkers for the Prediction of Fetal Ventricular Septal Defect. Front. Genet. 2021, 12, 717208. [Google Scholar] [CrossRef]
  85. Zhu, S.; Cao, L.; Zhu, J.; Kong, L.; Jin, J.; Qian, L.; Zhu, C.; Hu, X.; Li, M.; Guo, X.; et al. Identification of Maternal Serum MicroRNAs as Novel Non-Invasive Biomarkers for Prenatal Detection of Fetal Congenital Heart Defects. Clin. Chim. Acta 2013, 424, 66–72. [Google Scholar] [CrossRef] [PubMed]
  86. Gu, H.; Chen, L.; Xue, J.; Huang, T.; Wei, X.; Liu, D.; Ma, W.; Cao, S.; Yuan, Z. Expression Profile of Maternal Circulating MicroRNAs as Non-Invasive Biomarkers for Prenatal Diagnosis of Congenital Heart Defects. Biomed. Pharmacother. 2019, 109, 823–830. [Google Scholar] [CrossRef]
  87. Yang, Q.; Wu, F.; Mi, Y.; Wang, F.; Cai, K.; Yang, X.; Zhang, R.; Liu, L.; Zhang, Y.; Wang, Y.; et al. Aberrant Expression of MiR-29b-3p Influences Heart Development and Cardiomyocyte Proliferation by Targeting NOTCH2. Cell Prolif. 2020, 53, e12764. [Google Scholar] [CrossRef]
  88. Kong, D.; Li, H.; Ren, C.; Yang, W.; Zhang, Z. MicroRNAs in Fetal Umbilical Cord Blood as a Prenatal Screening Tool for Congenital Heart Disease. Ann. Clin. Lab. Sci. 2021, 51, 705–712. [Google Scholar]
  89. Kehler, L.; Biro, O.; Lazar, L.; Rigo, J.; Nagy, B. Elevated Hsa-MiR-99a Levels in Maternal Plasma May Indicate Congenital Heart Defects. Biomed. Rep. 2015, 3, 869–873. [Google Scholar] [CrossRef]
  90. Felekkis, K.; Papaneophytou, C. Challenges in Using Circulating Micro-RNAs as Biomarkers for Cardiovascular Diseases. Int. J. Mol. Sci. 2020, 21, 561. [Google Scholar] [CrossRef]
  91. Thiene, G.; Frescura, C. Anatomical and Pathophysiological Classification of Congenital Heart Disease. Cardiovasc. Pathol. 2010, 19, 259–274. [Google Scholar] [CrossRef]
  92. Ombelet, F.; Goossens, E.; Van De Bruaene, A.; Budts, W.; Moons, P. Newly Developed Adult Congenital Heart Disease Anatomic and Physiological Classification: First Predictive Validity Evaluation. J. Am. Heart Assoc. 2020, 9, e014988. [Google Scholar] [CrossRef]
  93. González-Moyotl, N.; Huesca-Gómez, C.; Torres-Paz, Y.E.; Fuentevilla-Álvarez, G.; Romero-Maldonado, S.; Sámano, R.; Soto, M.E.; Martínez-Rosas, M.; Domínguez-López, A.; Gamboa, R. Paediatrics Congenital Heart Disease Is Associated with Plasma MiRNAs. Pediatr. Res. 2024, 96, 1220–1227. [Google Scholar] [CrossRef]
  94. Khairy, P.; Poirier, N.; Mercier, L.-A. Univentricular Heart. Circulation 2007, 115, 800–812. [Google Scholar] [CrossRef]
  95. Kritzmire, S.M.; Thomas, A.; Horenstein, M.S.; Cossu, A.E. Hypoplastic Left Heart Syndrome; StatPearls: Treasure Island, FL, USA, 2025. [Google Scholar]
  96. Horenstein, M.S.; Diaz-Frias, J.; Guillaume, M. Tetralogy of Fallot; StatPearls: Treasure Island, FL, USA, 2024. [Google Scholar]
  97. van der Ven, J.P.G.; van den Bosch, E.; Bogers, A.J.C.C.; Helbing, W.A. Current Outcomes and Treatment of Tetralogy of Fallot. F1000Res 2019, 8, 1530. [Google Scholar] [CrossRef] [PubMed]
  98. O’Brien, J.E.; Kibiryeva, N.; Zhou, X.-G.; Marshall, J.A.; Lofland, G.K.; Artman, M.; Chen, J.; Bittel, D.C. Noncoding RNA Expression in Myocardium from Infants with Tetralogy of Fallot. Circ. Cardiovasc. Genet. 2012, 5, 279–286. [Google Scholar] [CrossRef]
  99. Bittel, D.; Kibiryeva, N.; Marshall, J.; O’Brien, J. MicroRNA-421 Dysregulation Is Associated with Tetralogy of Fallot. Cells 2014, 3, 713–723. [Google Scholar] [CrossRef]
  100. Zhang, J.; Chang, J.-J.; Xu, F.; Ma, X.-J.; Wu, Y.; Li, W.-C.; Wang, H.-J.; Huang, G.-Y.; Ma, D. MicroRNA Deregulation in Right Ventricular Outflow Tract Myocardium in Nonsyndromic Tetralogy of Fallot. Can. J. Cardiol. 2013, 29, 1695–1703. [Google Scholar] [CrossRef] [PubMed]
  101. Liang, D.; Xu, X.; Deng, F.; Feng, J.; Zhang, H.; Liu, Y.; Zhang, Y.; Pan, L.; Liu, Y.; Zhang, D.; et al. MiRNA-940 Reduction Contributes to Human Tetralogy of Fallot Development. J. Cell. Mol. Med. 2014, 18, 1830–1839. [Google Scholar] [CrossRef]
  102. Wu, Y.; Ma, X.-J.; Wang, H.-J.; Li, W.-C.; Chen, L.; Ma, D.; Huang, G.-Y. Expression of Cx43-Related MicroRNAs in Patients with Tetralogy of Fallot. World J. Pediatr. 2014, 10, 138–144. [Google Scholar] [CrossRef] [PubMed]
  103. Grunert, M.; Appelt, S.; Dunkel, I.; Berger, F.; Sperling, S.R. Altered MicroRNA and Target Gene Expression Related to Tetralogy of Fallot. Sci. Rep. 2019, 9, 19063. [Google Scholar] [CrossRef]
  104. Huesca-Gomez, C.; Gonzalez-Moyotl, N.; Romero-Maldonado, S.; Samano, R.; Gamboa, R. Role of Circulating MicroRNAs in Tetralogy of Fallot. Adv. Interv. Cardiol. 2025, 21, 247–254. [Google Scholar] [CrossRef] [PubMed]
  105. Yang, H.; Li, Y.; Chen, Q.; Li, S.; Yang, Y.; Lyu, G. Analyzing Exosomal MiRNA Profiles in Tetralogy of Fallot Fetuses’ Amniotic Fluid. Sci. Rep. 2025, 15, 96. [Google Scholar] [CrossRef] [PubMed]
  106. Bassett, A.S.; Costain, G.A.; Marshall, C.R.; Scherer, S.; Ogura, L.; Merico, D.; Silversides, C. Micrornas and Genetic Risk for Tetralogy of Fallot. Can. J. Cardiol. 2015, 31, S189–S190. [Google Scholar] [CrossRef]
  107. You, G.; Zu, B.; Wang, B.; Fu, Q.; Li, F. Identification of MiRNA–MRNA–TFs Regulatory Network and Crucial Pathways Involved in Tetralogy of Fallot. Front. Genet. 2020, 11, 552. [Google Scholar] [CrossRef] [PubMed]
  108. Wang, B.; Shi, G.; Zhu, Z.; Chen, H.; Fu, Q. Sexual Difference of Small RNA Expression in Tetralogy of Fallot. Sci. Rep. 2018, 8, 12847. [Google Scholar] [CrossRef]
  109. Menillo, A.M.; Alahmadi, M.H.; Pearson-Shaver, A.L. Atrial Septal Defect; StatPearls: Treasure Island, FL, USA, 2025. [Google Scholar]
  110. Alahmadi, M.H.; Oliver, T.I. Ventricular Septal Defect; StatPearls: Treasure Island, FL, USA, 2024. [Google Scholar]
  111. Li, D.; Ji, L.; Liu, L.; Liu, Y.; Hou, H.; Yu, K.; Sun, Q.; Zhao, Z. Characterization of Circulating MicroRNA Expression in Patients with a Ventricular Septal Defect. PLoS ONE 2014, 9, e106318. [Google Scholar] [CrossRef]
  112. Yu, K.; Ji, Y.; Wang, H.; Xuan, Q.K.; Li, B.B.; Xiao, J.J.; Sun, W.; Kong, X.Q. Association of MiR-196a2, MiR-27a, and MiR-499 Polymorphisms with Isolated Congenital Heart Disease in a Chinese Population. Genet. Mol. Res. 2016, 15, 8929. [Google Scholar] [CrossRef]
  113. Wang, Y.; Du, X.; Zhou, Z.; Jiang, J.; Zhang, Z.; Ye, L.; Hong, H. A Gain-of-Function ACTC1 3′UTR Mutation That Introduces a MiR-139-5p Target Site May Be Associated with a Dominant Familial Atrial Septal Defect. Sci. Rep. 2016, 6, 25404. [Google Scholar] [CrossRef]
  114. Han, S.; Wang, W.-J.; Duan, L.; Hou, Z.-L.; Zeng, J.-Y.; Li, L.; Meng, M.-Y.; Zhang, Y.-Y.; Wang, Y.; Xie, Y.-H.; et al. MicroRNA Profiling of Patients with Sporadic Atrial Septal Defect. Biotechnol. Biotechnol. Equip. 2019, 33, 510–519. [Google Scholar] [CrossRef]
  115. Jia, L.; Limeng, D.; Xiaoyin, T.; Junwen, W.; Xintong, Z.; Gang, X.; Yun, B.; Hong, G. A Novel Splicing Mutation c.335–1 G>A in the Cardiac Transcription Factor NKX2-5 Leads to Familial Atrial Septal Defect Through MiR-19 and PYK2. Stem Cell Rev. Rep. 2022, 18, 2646–2661. [Google Scholar] [CrossRef]
  116. Ahmed, I.; Alahmadi, M.H.; Anjum, F. Atrioventricular Septal Defect; StatPearls: Treasure Island, FL, USA, 2025. [Google Scholar]
  117. Coppola, A.; Romito, A.; Borel, C.; Gehrig, C.; Gagnebin, M.; Falconnet, E.; Izzo, A.; Altucci, L.; Banfi, S.; Antonarakis, S.E.; et al. Cardiomyogenesis Is Controlled by the MiR-99a/Let-7c Cluster and Epigenetic Modifications. Stem Cell Res. 2014, 12, 323–337. [Google Scholar] [CrossRef]
  118. Wang, L.; Li, Z.; Song, X.; Liu, L.; Su, G.; Cui, Y. Bioinformatic Analysis of Genes and MicroRNAs Associated with Atrioventricular Septal Defect in Down Syndrome Patients. Int. Heart J. 2016, 57, 490–495. [Google Scholar] [CrossRef] [PubMed]
  119. Smith, M.A.; Chiacchia, S.; Boehme, J.; Datar, S.A.; Morell, E.; Keller, R.L.; Romer, A.; Colglazier, E.; Parker, C.; Becerra, J.; et al. MicroRNA in Pediatric Pulmonary Hypertension MicroRNA Profiling to Inform Disease Classification, Severity, and Treatment Response in Pediatric Pulmonary Hypertension. Am. J. Physiol. Heart Circ. Physiol. 2025, 328, H47–H57. [Google Scholar] [CrossRef]
  120. Chouvarine, P.; Geldner, J.; Giagnorio, R.; Legchenko, E.; Bertram, H.; Hansmann, G. Trans-Right–Ventricle and Transpulmonary MicroRNA Gradients in Human Pulmonary Arterial Hypertension. Pediatr. Crit. Care Med. 2020, 21, 340–349. [Google Scholar] [CrossRef] [PubMed]
  121. Long, L.; Xiao, Y.; Yin, X.; Gao, S.; Zhou, L.; Liu, H. Expression of Serum MiR-27b and MiR-451 in Patients with Congenital Heart Disease Associated Pulmonary Artery Hypertension and Risk Factor Analysis. Exp. Ther. Med. 2020, 20, 3196–3202. [Google Scholar] [CrossRef]
  122. Deng, L.; Blanco, F.J.; Stevens, H.; Lu, R.; Caudrillier, A.; McBride, M.; McClure, J.D.; Grant, J.; Thomas, M.; Frid, M.; et al. MicroRNA-143 Activation Regulates Smooth Muscle and Endothelial Cell Crosstalk in Pulmonary Arterial Hypertension. Circ. Res. 2015, 117, 870–883. [Google Scholar] [CrossRef] [PubMed]
  123. Zhang, Y.-Z.; Li, G.; Yan, D.-L.; Liu, Y.-L. MicroRNA-98 Can Serve as a Diagnostic Marker for Congenital Heart Disease-Associated Pulmonary Artery Hypertension. Chin. Med. J. 2020, 133, 3007–3009. [Google Scholar] [CrossRef]
  124. Su, D.; Huang, Y.; Liu, D.; Huang, Y.; Ye, B.; Qin, S.; Chen, C.; Pang, Y. Bioinformatic Analysis of Dysregulated Circular RNAs in Pediatric Pulmonary Hypertension Linked Congenital Heart Disease. Transl. Pediatr. 2022, 11, 715–727. [Google Scholar] [CrossRef]
  125. Ma, K.; Zhao, Q.; Chen, W.; Zhang, H.; Li, S.; Pan, X.; Chen, Q. Human Lung MicroRNA Profiling in Pulmonary Arterial Hypertension Secondary to Congenital Heart Defect. Pediatr. Pulmonol. 2015, 50, 1214–1223. [Google Scholar] [CrossRef]
  126. Estephan, L.E.; Genuardi, M.V.; Kosanovich, C.M.; Risbano, M.G.; Zhang, Y.; Petro, N.; Watson, A.; Al Aaraj, Y.; Sembrat, J.C.; Rojas, M.; et al. Distinct Plasma Gradients of MicroRNA-204 in the Pulmonary Circulation of Patients Suffering from WHO Groups I and II Pulmonary Hypertension. Pulm. Circ. 2019, 9, 2045894019840646. [Google Scholar] [CrossRef]
  127. Katsaros, O.; Ktenopoulos, N.; Korovesis, T.; Benetos, G.; Apostolos, A.; Koliastasis, L.; Sagris, M.; Milaras, N.; Latsios, G.; Synetos, A.; et al. Bicuspid Aortic Valve Disease: From Pathophysiology to Treatment. J. Clin. Med. 2024, 13, 4970. [Google Scholar] [CrossRef]
  128. Siu, S.C.; Silversides, C.K. Bicuspid Aortic Valve Disease. J. Am. Coll. Cardiol. 2010, 55, 2789–2800. [Google Scholar] [CrossRef]
  129. Antequera-González, B.; Collell-Hernández, R.; Martínez-Micaelo, N.; Marimon-Blanch, C.; Carbonell-Prat, B.; Escribano, J.; Alegret, J.M. MiR-130a Expression Is Related to Aortic Dilation in Bicuspid Aortic Valve Children. Pediatr. Res. 2024, 95, 1741–1748. [Google Scholar] [CrossRef] [PubMed]
  130. Sanchez-Garcia, A.d.J.; Soule-Egea, M.; Fuentevilla-Alvarez, G.; Vargas-Alarcon, G.; Hernández-Mejia, B.I.; Martínez-Hernández, H.; Mora-Canela, S.L.; Santibanez-Escobar, F.; Ávila-Martinez, V.; Castrejón-Tellez, V.; et al. Role of MiRNAs in Regulating Ascending Aortic Dilation in Bicuspid Aortic Valve Patients Operated for Aortic Stenosis. Int. J. Mol. Sci. 2025, 26, 779. [Google Scholar] [CrossRef]
  131. Naito, S.; Sequeira-Gross, T.; Petersen, J.; Detlef, I.; Sachse, M.; Zeller, T.; Reichenspurner, H.; Girdauskas, E. Circulating MicroRNAs in the Prediction of BAV Aortopathy: Do the Expression Patterns Correlate between Blood and Aortic Tissue? Rev. Cardiovasc. Med. 2022, 23, 47. [Google Scholar] [CrossRef]
  132. Borghini, A.; Foffa, I.; Pulignani, S.; Vecoli, C.; Ait-Ali, L.; Andreassi, M.G. MiRNome Profiling in Bicuspid Aortic Valve-Associated Aortopathy by Next-Generation Sequencing. Int. J. Mol. Sci. 2017, 18, 2498. [Google Scholar] [CrossRef] [PubMed]
  133. Sophocleous, F.; De Garate, E.; Bigotti, M.G.; Anwar, M.; Jover, E.; Chamorro-Jorganes, A.; Rajakaruna, C.; Mitrousi, K.; De Francesco, V.; Wilson, A.; et al. A Segmental Approach from Molecular Profiling to Medical Imaging to Study Bicuspid Aortic Valve Aortopathy. Cells 2022, 11, 3721. [Google Scholar] [CrossRef] [PubMed]
  134. Nigam, V.; Sievers, H.H.; Jensen, B.C.; Sier, H.A.; Simpson, P.C.; Srivastava, D.; Mohamed, S.A. Altered MicroRNAs in Bicuspid Aortic Valve: A Comparison between Stenotic and Insufficient Valves. J. Heart Valve Dis. 2010, 19, 459–465. [Google Scholar]
  135. Ragusa, R.; Di Molfetta, A.; D’Aurizio, R.; Del Turco, S.; Cabiati, M.; Del Ry, S.; Basta, G.; Pitto, L.; Amodeo, A.; Trivella, M.G.; et al. Variations of Circulating MiRNA in Paediatric Patients with Heart Failure Supported with Ventricular Assist Device: A Pilot Study. Sci. Rep. 2020, 10, 5905. [Google Scholar] [CrossRef]
  136. Klimczak-Tomaniak, D.; Haponiuk-Skwarlińska, J.; Kuch, M.; Pączek, L. Crosstalk between MicroRNA and Oxidative Stress in Heart Failure: A Systematic Review. Int. J. Mol. Sci. 2022, 23, 15013. [Google Scholar] [CrossRef]
  137. Toni, L.; Hailu, F.; Sucharov, C.C. Dysregulated Micro-RNAs and Long Noncoding RNAs in Cardiac Development and Pediatric Heart Failure. Am. J. Physiol.-Heart Circ. Physiol. 2020, 318, H1308–H1315. [Google Scholar] [CrossRef]
  138. Stauffer, B.L.; Russell, G.; Nunley, K.; Miyamoto, S.D.; Sucharov, C.C. MiRNA Expression in Pediatric Failing Human Heart. J. Mol. Cell. Cardiol. 2013, 57, 43–46. [Google Scholar] [CrossRef]
  139. Su, X.; Liang, H.; Wang, H.; Chen, G.; Jiang, H.; Wu, Q.; Liu, T.; Liu, Q.; Yu, T.; Gu, Y.; et al. Over-Expression of MicroRNA-1 Causes Arrhythmia by Disturbing Intracellular Trafficking System. Sci. Rep. 2017, 7, 46259. [Google Scholar] [CrossRef] [PubMed]
  140. Zhang, Y.; Sun, L.; Zhang, Y.; Liang, H.; Li, X.; Cai, R.; Wang, L.; Du, W.; Zhang, R.; Li, J.; et al. Overexpression of MicroRNA-1 Causes Atrioventricular Block in Rodents. Int. J. Biol. Sci. 2013, 9, 455–462. [Google Scholar] [CrossRef]
  141. Moric-Janiszewska, E.; Smolik, S.; Morka, A.; Szydłowski, L.; Kapral, M. Expression Levels of Serum Circulating MicroRNAs in Pediatric Patients with Ventricular and Supraventricular Arrhythmias. Adv. Med. Sci. 2021, 66, 411–417. [Google Scholar] [CrossRef]
  142. Sun, L.; Sun, S.; Zeng, S.; Li, Y.; Pan, W.; Zhang, Z. Expression of Circulating MicroRNA-1 and MicroRNA-133 in Pediatric Patients with Tachycardia. Mol. Med. Rep. 2015, 11, 4039–4046. [Google Scholar] [CrossRef]
  143. Al-Akchar, M.; Siddique, M.S. Long QT Syndrome; StatPearls: Treasure Island, FL, USA, 2025. [Google Scholar]
  144. Matkovich, S.J.; Wang, W.; Tu, Y.; Eschenbacher, W.H.; Dorn, L.E.; Condorelli, G.; Diwan, A.; Nerbonne, J.M.; Dorn, G.W. MicroRNA-133a Protects Against Myocardial Fibrosis and Modulates Electrical Repolarization Without Affecting Hypertrophy in Pressure-Overloaded Adult Hearts. Circ. Res. 2010, 106, 166–175. [Google Scholar] [CrossRef] [PubMed]
  145. Shan, H.; Zhang, Y.; Lu, Y.; Zhang, Y.; Pan, Z.; Cai, B.; Wang, N.; Li, X.; Feng, T.; Hong, Y.; et al. Downregulation of MiR-133 and MiR-590 Contributes to Nicotine-Induced Atrial Remodelling in Canines. Cardiovasc. Res. 2009, 83, 465–472. [Google Scholar] [CrossRef]
  146. Xiao, L.; Xiao, J.; Luo, X.; Lin, H.; Wang, Z.; Nattel, S. Feedback Remodeling of Cardiac Potassium Current Expression. Circulation 2008, 118, 983–992. [Google Scholar] [CrossRef] [PubMed]
  147. Hedley, P.L.; Jørgensen, P.; Schlamowitz, S.; Wangari, R.; Moolman-Smook, J.; Brink, P.A.; Kanters, J.K.; Corfield, V.A.; Christiansen, M. The Genetic Basis of Long QT and Short QT Syndromes: A Mutation Update. Hum. Mutat. 2009, 30, 1486–1511. [Google Scholar] [CrossRef]
  148. Hedley, P.L.; Carlsen, A.L.; Christiansen, K.M.; Kanters, J.K.; Behr, E.R.; Corfield, V.A.; Christiansen, M. MicroRNAs in Cardiac Arrhythmia: DNA Sequence Variation of MiR-1 and MiR-133A in Long QT Syndrome. Scand. J. Clin. Lab. Investig. 2014, 74, 485–491. [Google Scholar] [CrossRef]
  149. Vardas, E.P.; Oikonomou, E.; Vardas, P.E.; Tousoulis, D. MicroRNAs as Prognostic Biomarkers for Atrial Fibrillation Recurrence After Catheter Ablation: Current Evidence and Future Directions. Biomedicines 2024, 13, 32. [Google Scholar] [CrossRef] [PubMed]
  150. Kiliszek, M.; Maciak, K.; Maciejak, A.; Krzyżanowski, K.; Wierzbowski, R.; Gora, M.; Burzynska, B.; Segiet, A.; Skrobowski, A. Serum MicroRNA in Patients Undergoing Atrial Fibrillation Ablation. Sci. Rep. 2020, 10, 4424. [Google Scholar] [CrossRef] [PubMed]
  151. Hailu, F.T.; Karimpour-Fard, A.; Neltner, B.; Stauffer, B.L.; Lipshultz, S.; Miyamoto, S.D.; Sucharov, C.C. Circulating and Cardiac Tissue MiRNAs in Children with Dilated Cardiomyopathy. J. Cardiovasc. Dev. Dis. 2023, 10, 391. [Google Scholar] [CrossRef]
  152. Hirai, K.; Ousaka, D.; Fukushima, Y.; Kondo, M.; Eitoku, T.; Shigemitsu, Y.; Hara, M.; Baba, K.; Iwasaki, T.; Kasahara, S.; et al. Cardiosphere-Derived Exosomal MicroRNAs for Myocardial Repair in Pediatric Dilated Cardiomyopathy. Sci. Transl. Med. 2020, 12, eabb3336. [Google Scholar] [CrossRef]
  153. Jiao, M.; You, H.-Z.; Yang, X.-Y.; Yuan, H.; Li, Y.-L.; Liu, W.-X.; Jin, M.; Du, J. Circulating MicroRNA Signature for the Diagnosis of Childhood Dilated Cardiomyopathy. Sci. Rep. 2018, 8, 724. [Google Scholar] [CrossRef] [PubMed]
  154. Enes Coşkun, M.; Kervancıoğlu, M.; Öztuzcu, S.; Yılmaz Coşkun, F.; Ergün, S.; Başpınar, O.; Kılınç, M.; Temel, L.; Coşkun, M.Y. Plasma MicroRNA Profiling of Children with Idiopathic Dilated Cardiomyopathy. Biomarkers 2016, 21, 56–61. [Google Scholar] [CrossRef]
  155. Meng, Z.; Lu, M. RNA Interference-Induced Innate Immunity, Off-Target Effect, or Immune Adjuvant? Front. Immunol. 2017, 8, 331. [Google Scholar] [CrossRef]
  156. Zhang, S.; Cheng, Z.; Wang, Y.; Han, T. The Risks of MiRNA Therapeutics: In a Drug Target Perspective. Drug Des. Dev. Ther. 2021, 15, 721–733. [Google Scholar] [CrossRef]
  157. Segal, M.; Slack, F.J. Challenges Identifying Efficacious MiRNA Therapeutics for Cancer. Expert. Opin. Drug Discov. 2020, 15, 987–991. [Google Scholar] [CrossRef]
  158. Haupenthal, J.; Baehr, C.; Kiermayer, S.; Zeuzem, S.; Piiper, A. Inhibition of RNAse A Family Enzymes Prevents Degradation and Loss of Silencing Activity of SiRNAs in Serum. Biochem. Pharmacol. 2006, 71, 702–710. [Google Scholar] [CrossRef]
  159. Segal, M.; Biscans, A.; Gilles, M.-E.; Anastasiadou, E.; De Luca, R.; Lim, J.; Khvorova, A.; Slack, F.J. Hydrophobically Modified Let-7b MiRNA Enhances Biodistribution to NSCLC and Downregulates HMGA2 In Vivo. Mol. Ther. Nucleic Acids 2020, 19, 267–277. [Google Scholar] [CrossRef]
  160. Scheideler, M.; Vidakovic, I.; Prassl, R. Lipid Nanocarriers for MicroRNA Delivery. Chem. Phys. Lipids 2020, 226, 104837. [Google Scholar] [CrossRef]
  161. Akbarzadeh, A.; Rezaei-Sadabady, R.; Davaran, S.; Joo, S.W.; Zarghami, N.; Hanifehpour, Y.; Samiei, M.; Kouhi, M.; Nejati-Koshki, K. Liposome: Classification, Preparation, and Applications. Nanoscale Res. Lett. 2013, 8, 102. [Google Scholar] [CrossRef]
  162. Gao, S.; Tian, H.; Guo, Y.; Li, Y.; Guo, Z.; Zhu, X.; Chen, X. MiRNA Oligonucleotide and Sponge for MiRNA-21 Inhibition Mediated by PEI-PLL in Breast Cancer Therapy. Acta Biomater. 2015, 25, 184–193. [Google Scholar] [CrossRef]
  163. Biray Avcı, Ç.; Özcan, İ.; Balcı, T.; Özer, Ö.; Gündüz, C. Design of Polyethylene Glycol–Polyethylenimine Nanocomplexes as Non-viral Carriers: Mir-150 Delivery to Chronic Myeloid Leukemia Cells. Cell Biol. Int. 2013, 37, 1205–1214. [Google Scholar] [CrossRef] [PubMed]
  164. Song, Y.; Zhang, C.; Zhang, J.; Jiao, Z.; Dong, N.; Wang, G.; Wang, Z.; Wang, L. Localized Injection of MiRNA-21-Enriched Extracellular Vesicles Effectively Restores Cardiac Function after Myocardial Infarction. Theranostics 2019, 9, 2346–2360. [Google Scholar] [CrossRef] [PubMed]
  165. Li, X.; Corbett, A.L.; Taatizadeh, E.; Tasnim, N.; Little, J.P.; Garnis, C.; Daugaard, M.; Guns, E.; Hoorfar, M.; Li, I.T.S. Challenges and Opportunities in Exosome Research—Perspectives from Biology, Engineering, and Cancer Therapy. APL Bioeng. 2019, 3, 011503. [Google Scholar] [CrossRef] [PubMed]
  166. Zhang, H.; Zhan, Q.; Huang, B.; Wang, Y.; Wang, X. AAV-Mediated Gene Therapy: Advancing Cardiovascular Disease Treatment. Front. Cardiovasc. Med. 2022, 9, 952755. [Google Scholar] [CrossRef]
  167. Yan, H.; Wang, H.; Zhu, X.; Huang, J.; Li, Y.; Zhou, K.; Hua, Y.; Yan, F.; Wang, D.-Z.; Luo, Y. Adeno-Associated Virus-Mediated Delivery of Anti-MiR-199a Tough Decoys Attenuates Cardiac Hypertrophy by Targeting PGC-1alpha. Mol. Ther. Nucleic Acids 2021, 23, 406–417. [Google Scholar] [CrossRef] [PubMed]
  168. Jaski, B.E.; Jessup, M.L.; Mancini, D.M.; Cappola, T.P.; Pauly, D.F.; Greenberg, B.; Borow, K.; Dittrich, H.; Zsebo, K.M.; Hajjar, R.J. Calcium Upregulation by Percutaneous Administration of Gene Therapy in Cardiac Disease (CUPID Trial), a First-in-Human Phase 1/2 Clinical Trial. J. Card. Fail. 2009, 15, 171–181. [Google Scholar] [CrossRef]
  169. Kopechek, J.A.; McTiernan, C.F.; Chen, X.; Zhu, J.; Mburu, M.; Feroze, R.; Whitehurst, D.A.; Lavery, L.; Cyriac, J.; Villanueva, F.S. Ultrasound and Microbubble-Targeted Delivery of a MicroRNA Inhibitor to the Heart Suppresses Cardiac Hypertrophy and Preserves Cardiac Function. Theranostics 2019, 9, 7088–7098. [Google Scholar] [CrossRef]
  170. Yang, H.; Qin, X.; Wang, H.; Zhao, X.; Liu, Y.; Wo, H.-T.; Liu, C.; Nishiga, M.; Chen, H.; Ge, J.; et al. An in Vivo MiRNA Delivery System for Restoring Infarcted Myocardium. ACS Nano 2019, 13, 9880–9894. [Google Scholar] [CrossRef]
  171. Fisher, R.K.; Mattern-Schain, S.I.; Best, M.D.; Kirkpatrick, S.S.; Freeman, M.B.; Grandas, O.H.; Mountain, D.J.H. Improving the Efficacy of Liposome-Mediated Vascular Gene Therapy via Lipid Surface Modifications. J. Surg. Res. 2017, 219, 136–144. [Google Scholar] [CrossRef]
  172. Wu, X.; Zhang, L.; Tong, X.; Wang, Y.; Zhang, Z.; Kong, X.; Ni, S.; Luo, X.; Zheng, M.; Tang, Y.; et al. MiCGR: Interpretable Deep Neural Network for Predicting Both Site-Level and Gene-Level Functional Targets of MicroRNA. Brief. Bioinform. 2024, 26, bbae616. [Google Scholar] [CrossRef]
  173. Errington, N.; Iremonger, J.; Pickworth, J.A.; Kariotis, S.; Rhodes, C.J.; Rothman, A.M.; Condliffe, R.; Elliot, C.A.; Kiely, D.G.; Howard, L.S.; et al. A Diagnostic MiRNA Signature for Pulmonary Arterial Hypertension Using a Consensus Machine Learning Approach. EBioMedicine 2021, 69, 103444. [Google Scholar] [CrossRef]
  174. Paolini, A.; Baldassarre, A.; Bruno, S.P.; Felli, C.; Muzi, C.; Ahmadi Badi, S.; Siadat, S.D.; Sarshar, M.; Masotti, A. Improving the Diagnostic Potential of Extracellular MiRNAs Coupled to Multiomics Data by Exploiting the Power of Artificial Intelligence. Front. Microbiol. 2022, 13, 888414. [Google Scholar] [CrossRef] [PubMed]
  175. White, A.K.; VanInsberghe, M.; Petriv, O.I.; Hamidi, M.; Sikorski, D.; Marra, M.A.; Piret, J.; Aparicio, S.; Hansen, C.L. High-Throughput Microfluidic Single-Cell RT-QPCR. Proc. Natl. Acad. Sci. USA 2011, 108, 13999–14004. [Google Scholar] [CrossRef] [PubMed]
  176. Faridani, O.R.; Abdullayev, I.; Hagemann-Jensen, M.; Schell, J.P.; Lanner, F.; Sandberg, R. Single-Cell Sequencing of the Small-RNA Transcriptome. Nat. Biotechnol. 2016, 34, 1264–1266. [Google Scholar] [CrossRef]
  177. Herbst, E.; Mandel-Gutfreund, Y.; Yakhini, Z.; Biran, H. Inferring Single-Cell and Spatial MicroRNA Activity from Transcriptomics Data. Commun. Biol. 2025, 8, 87. [Google Scholar] [CrossRef] [PubMed]
Figure 1. miRNA biogenesis and mechanisms of action.
Figure 1. miRNA biogenesis and mechanisms of action.
Jcm 14 06833 g001
Table 2. Key microRNAs in congenital heart diseases. ASD—atrial septal defect, BAV—bicuspid aortic valve, HF—heart failure, HLHS—hypoplastic left heart syndrome, TAV—tricuspid aortic valve, TOF—tetralogy of Fallot, UAV—unicuspid aortic valve, RVOT—right ventricle outflow tract.
Table 2. Key microRNAs in congenital heart diseases. ASD—atrial septal defect, BAV—bicuspid aortic valve, HF—heart failure, HLHS—hypoplastic left heart syndrome, TAV—tricuspid aortic valve, TOF—tetralogy of Fallot, UAV—unicuspid aortic valve, RVOT—right ventricle outflow tract.
DiseaseKey miRNAsExpression PatternClinical Significance and FunctionReferences
HLHSmiR-150-5pDownregulated in serumPotential biomarker for HF, especially when combined with NT-proBNP[7]
miR-129-5pDownregulated in serumPotential biomarker for HF, likely associated with hypoxic response[8]
TOFmiR-19b, miR-19c,
miR-375, miR-22
Upregulated in maternal serumPotential biomarker[85]
miR-421, miR-1201Upregulated in RV tissueMediator of RV remodeling in TOF, miR-421, negatively correlated with SOX4[98,99]
miR625-5p, miR-183-5pUpregulated in serumPotential biomarker[6]
miR-181d-5p, miR-142-5p, miR-339-5pDownregulated in serumPotential biomarker[6]
miR-424, miR-222Upregulated in RVOT tissueEnhanced proliferation, additionally, miR-222 leads to inhibition of cardiomyocyte differentiation (murine models)[100]
miR-940Downregulated in RVOT tissueEnhanced proliferation and inhibition lead to reduced migration, acting through the inhibition of JARID2, a protein essential for outflow tract morphogenesis[101]
miR-1, miR-206Downregulated in RVOT tissuesImparing myocardial conduction—increased expression of Cx-43, disrupted expression of KCNJ2 and TNNI1[6,102,103]
miR-221-5p, miR-21-5p, miR-155-5pUpregulated in serumPotential biomarkers, mainly active in hypoxia-sensitive signaling pathways[104]
miR-21-5p, miR-144-3pDownregulated in serum
miR-200a-3p, miR-10a-5pUpregulated in amniotic fluidPotential biomarkers, acting through inhibition of cardiomyocyte differentiation via suppression of the transcription factor GATA4, while miR-10a-5p additionally suppresses TBX5 and NKX2.5[105]
VSDmiR-1-1Downregulated in heart tissueInhibition of GJA1 and SOX9[10]
miR-181cUpregulated in heart tissueDownregulation of BMPR2[10]
hsa-let-7e-5p, hsa-miR-155-5p, hsa-miR-222-3p, hsa-miR-379-5p, hsa-miR- 409-3p, hsa-miR-433, hsa-miR-487bDownregulated in serumImpact on the expression of genes critical for cardiac development, including HAND1, NKX2-4, TBX1, MAP2K4, GATA4, NOTCH1
and ZFPM2
[111]
hsa-miR-498Upregulated in serum
miR-142-5p, miR-4666a-3pDownregulated in maternal serumPotential biomarkers[86]
miR-1275, miR-3664-3pUpregulated in maternal serum
hsa-miR-146a-5pDownregulated in maternal serumPotential biomarker, acting through the suppression of NUMB expression, a negative regulator of the Notch pathway[84]
ASDmiR-196a2Collected from serumSNP of this miRNA is associated with disease susceptibility[112]
miR-139-5pMutation identified by whole-genome sequencingA mutation in the 3′UTR of the ACTC1 gene results in the de novo creation of a binding site for miR-139-5p and disease development[113]
hsa-let-7a, hsa-let-7b, hsa-miR-486Upregulated in serum, hsa-let-7a also showed a positive correlation between levels in maternal and child serumPotential biomarker in prenatal screening, miR-486 also exhibited significantly higher levels among patients with VSD and AVSD[11]
miR-29, miR-143/145Upregulated in murine atrial septum tissueInvolvement in the regulation of multiple pathways essential for morphogenesis.[114]
miR-17-92, miR-106b-25, miR-503/424Downregulated in murine atrial septum tissue
BAVmiR-130aDownregulated in serumPotential biomarker, levels reduced in patients with BAV and dilated aorta, acting through modulation of the TGF-β pathway[129]
let-7e-5p, miR-196a-5pUpregulated in aortic wall tissueChanges in concentration relative to controls were particularly pronounced in groups with BAV and aortic dilation[130]
miR-17a-5pDownregulated in aortic wall tissue
miR-21, miR-133a, miR-143, miR-145Positive correlation between plasma and aortic tissue concentrations among BAV and UAV patientsAssociation between investigated miRNAs and aortopathies[131]
miR-424-3p, miR-3688-3pDownregulated in aortic aneurysm tissue among patients with BAV compared to TAVModulation of the expression of genes such as Hippo, ErbB, and TGF-β involved in the development of the valvular apparatus[132]
miR-128-3pDownregulated in the dilated segments of aortic tissueAssociated with inflammatory response pathways[133]
miR-150-5p, and miR-199b-5pUpregulated in the dilated segments of aortic tissueRelated to eicosanoid synthesis and the regulation of VEGF signaling pathways
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kozik, A.; Piotrowski, M.; Karpierz, J.I.; Kowalewski, M.; Batko, J. The Hidden Regulators: MicroRNAs in Pediatric Heart Development and Disease. J. Clin. Med. 2025, 14, 6833. https://doi.org/10.3390/jcm14196833

AMA Style

Kozik A, Piotrowski M, Karpierz JI, Kowalewski M, Batko J. The Hidden Regulators: MicroRNAs in Pediatric Heart Development and Disease. Journal of Clinical Medicine. 2025; 14(19):6833. https://doi.org/10.3390/jcm14196833

Chicago/Turabian Style

Kozik, Adam, Michał Piotrowski, Julia Izabela Karpierz, Mariusz Kowalewski, and Jakub Batko. 2025. "The Hidden Regulators: MicroRNAs in Pediatric Heart Development and Disease" Journal of Clinical Medicine 14, no. 19: 6833. https://doi.org/10.3390/jcm14196833

APA Style

Kozik, A., Piotrowski, M., Karpierz, J. I., Kowalewski, M., & Batko, J. (2025). The Hidden Regulators: MicroRNAs in Pediatric Heart Development and Disease. Journal of Clinical Medicine, 14(19), 6833. https://doi.org/10.3390/jcm14196833

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop