Advertisement
Review Article|Articles in Press

Role of microRNAs in arrhythmogenic cardiomyopathy: translation as biomarkers into clinical practice

  • Author Footnotes
    & These authors equally contributed as co-first authors.
    Mireia Alcalde
    Footnotes
    & These authors equally contributed as co-first authors.
    Affiliations
    Cardiovascular Genetics Center, University of Girona-IDIBGI, Girona, Spain

    Centro de Investigación Biomédica en Red. Enfermedades Cardiovasculares, Madrid, Spain
    Search for articles by this author
  • Author Footnotes
    & These authors equally contributed as co-first authors.
    ,
    Author Footnotes
    # These authors equally contributed as corresponding authors.
    Rocío Toro
    Correspondence
    Corresponding author: Rocio Toro, Medicine Department, School of Medicine, 11003 Cádiz, Spain
    Footnotes
    & These authors equally contributed as co-first authors.
    # These authors equally contributed as corresponding authors.
    Affiliations
    Medicine Department, School of Medicine, Cadiz, Spain

    Research Unit, Biomedical Research and Innovation Institute of Cadiz (INiBICA), Puerta del Mar University Hospital, Cádiz, Spain
    Search for articles by this author
  • Fernando Bonet
    Affiliations
    Medicine Department, School of Medicine, Cadiz, Spain

    Research Unit, Biomedical Research and Innovation Institute of Cadiz (INiBICA), Puerta del Mar University Hospital, Cádiz, Spain
    Search for articles by this author
  • José Córdoba-Caballero
    Affiliations
    Medicine Department, School of Medicine, Cadiz, Spain

    Research Unit, Biomedical Research and Innovation Institute of Cadiz (INiBICA), Puerta del Mar University Hospital, Cádiz, Spain
    Search for articles by this author
  • Estefanía Martínez-Barrios
    Affiliations
    Pediatric Arrhythmias, Inherited Cardiac Diseases and Sudden Death Unit, Cardiology Department, Sant Joan de Déu Hospital, Barcelona, Spain

    European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart), Amsterdam, the Netherlands

    Arrítmies Pediàtriques, Cardiologia Genètica i Mort Sobtada, Malalties Cardiovasculars en el Desenvolupament, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
    Search for articles by this author
  • Juan Antonio Ranea
    Affiliations
    Departamento de Biología Molecular y Bioquímica, Universidad de Málaga, Málaga, Spain

    Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain

    Centro de Investigación Biomedica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
    Search for articles by this author
  • Marta Vallverdú-Prats
    Affiliations
    Cardiovascular Genetics Center, University of Girona-IDIBGI, Girona, Spain

    Centro de Investigación Biomédica en Red. Enfermedades Cardiovasculares, Madrid, Spain
    Search for articles by this author
  • Ramon Brugada
    Affiliations
    Cardiovascular Genetics Center, University of Girona-IDIBGI, Girona, Spain

    Centro de Investigación Biomédica en Red. Enfermedades Cardiovasculares, Madrid, Spain

    Medical Science Department, School of Medicine, University of Girona, Girona, Spain

    Cardiology Department, Hospital Josep Trueta, Girona, Spain
    Search for articles by this author
  • Viviana Meraviglia
    Affiliations
    Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands
    Search for articles by this author
  • Milena Bellin
    Affiliations
    Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands

    Department of Biology, University of Padua, Padua, Italy

    Veneto Institute of Molecular Medicine, Padua, Italy
    Search for articles by this author
  • Georgia Sarquella-Brugada
    Affiliations
    Pediatric Arrhythmias, Inherited Cardiac Diseases and Sudden Death Unit, Cardiology Department, Sant Joan de Déu Hospital, Barcelona, Spain

    European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart), Amsterdam, the Netherlands

    Arrítmies Pediàtriques, Cardiologia Genètica i Mort Sobtada, Malalties Cardiovasculars en el Desenvolupament, Institut de Recerca Sant Joan de Déu, Barcelona, Spain

    Medical Science Department, School of Medicine, University of Girona, Girona, Spain
    Search for articles by this author
  • Author Footnotes
    # These authors equally contributed as corresponding authors.
    Oscar Campuzano
    Footnotes
    # These authors equally contributed as corresponding authors.
    Affiliations
    Cardiovascular Genetics Center, University of Girona-IDIBGI, Girona, Spain

    Centro de Investigación Biomédica en Red. Enfermedades Cardiovasculares, Madrid, Spain

    Medical Science Department, School of Medicine, University of Girona, Girona, Spain
    Search for articles by this author
  • Author Footnotes
    & These authors equally contributed as co-first authors.
    # These authors equally contributed as corresponding authors.
Open AccessPublished:April 24, 2023DOI:https://doi.org/10.1016/j.trsl.2023.04.003

      ABSTRACT

      Arrhythmogenic cardiomyopathy is a rare inherited entity, characterized by a progressive fibro-fatty replacement of the myocardium. It leads to malignant arrhythmias and a high risk of sudden cardiac death. Incomplete penetrance and variable expressivity are hallmarks of this arrhythmogenic cardiac disease, where the first manifestation may be syncope and sudden cardiac death, often triggered by physical exercise. Early identification of individuals at risk is crucial to adopt protective and ideally personalized measures to prevent lethal episodes. The genetic analysis identifies deleterious rare variants in nearly 70% of cases, mostly in genes encoding proteins of the desmosome. However, other factors may modulate the phenotype onset and outcome of disease, such as microRNAs. These small noncoding RNAs play a key role in gene expression regulation and the network of cellular processes. In recent years, data focused on the role of microRNAs as potential biomarkers in arrhythmogenic cardiomyopathy have progressively increased. A better understanding of the functions and interactions of microRNAs will likely have clinical implications. Herein, we propose an exhaustive review of the literature regarding these noncoding RNAs, their versatile mechanisms of gene regulation and present novel targets in arrhythmogenic cardiomyopathy.

      Keywords

      Abbreviations:

      ACM (arrhythmogenic cardiomyopathy), BIN1 (bridging integrator 1), CDH2 (cadherin 2), cTNI (troponin), CTNNA3 (T-catenin), DES (desmin), DSG2 (desmoglein-2), DSP (desmoplakin), FLNC (filamin C), GAL-3 (galetin-3), HSP70 (heat shock protein 70), IVT (idiopathic ventricular tachycardia), JUP (plakoglobin), LMNA (lamin A/C), miRNA (microRNA), mRNA (messenger RNA), ncRNA (noncoding RNA), NGS (next-generation sequencing), PLN (phospholamban), PPKP2 (plakophilin-2), RV (right ventricle), SCD (sudden cardiac death), SCN5A (sodium voltage-gated channel alpha subunit 5), TGFβ (transforming growth factor β), TMEM43 (transmembrane protein 43), TTN (titin), VF (ventricular fibrillation), VT (ventricular tachycardia), VUS (variant unknown significance)

      ARRHYTHMOGENIC CARDIOMYOPATHY

      Arrhythmogenic cardiomyopathy (ACM) includes the classical abnormality of the right ventricle (RV) in which the myocardium is progressively substituted by fibro-fatty tissue,
      • Kohela A
      • van Rooij E.
      Fibro-fatty remodelling in arrhythmogenic cardiomyopathy.
      named firstly arrhythmogenic right ventricular dysplasia and later arrhythmogenic right ventricular cardiomyopathy. In recent years, solely left ventricular and biventricular phenotypes have been also reported. This progressive tissue substitution leads to palpitations, syncope and/or cardiac arrest, secondary to ventricular tachycardia (VT) or fibrillation (VF).
      • Wilde AAM
      • Semsarian C
      • Marquez MF
      • et al.
      European Heart Rhythm Association (EHRA)/Heart Rhythm Society (HRS)/Asia Pacific Heart Rhythm Society (APHRS)/Latin American Heart Rhythm Society (LAHRS) expert consensus statement on the state of genetic testing for cardiac diseases.
      Sudden cardiac death (SCD) may be the first manifestation of the disease, occurring even in the absence of an evident structural alteration of the myocardium, as a consequence of electrical disruptions that lead to malignant arrhythmia.
      • Grassi S
      • Campuzano O
      • Coll M
      • et al.
      Update on the diagnostic pitfalls of autopsy and post-mortem genetic testing in cardiomyopathies.
      This occurs in the early stages of the disease due to protein conformational changes without a high degree of fibro-fatty replacement of cardiomyocytes. SCD episodes usually occur in young men, with physical exercise being the main trigger of the lethal arrhythmias in genetically predisposed hearts.
      • D'Ascenzi F
      • Valentini F
      • Pistoresi S
      • et al.
      Causes of sudden cardiac death in young athletes and non-athletes: systematic review and meta-analysis: sudden cardiac death in the young.
      ACM is a rare arrhythmogenic entity with an estimated prevalence between 1:2000 and 1: 5000 (OMIM #107970; ORPHA247), depending on the population. ACM is an inherited disorder, characterized by incomplete penetrance and variable phenotype expressivity.
      • Krahn AD
      • Wilde AAM
      • Calkins H
      • et al.
      Arrhythmogenic right ventricular cardiomyopathy.
      The pathophysiological mechanisms that underpin this diverse and pleiotropic disease remain poorly characterized.
      • Gerull B
      • Brodehl A.
      Insights into genetics and pathophysiology of arrhythmogenic cardiomyopathy.
      Therefore, relatives of a diagnosed patient who carry the same deleterious variant may be at risk of malignant arrhythmias, despite being asymptomatic. ACM affects adolescents infrequently and children rarely, despite clinical assessment.
      • Cohen MI
      • Atkins MB.
      Arrhythmogenic right ventricular cardiomyopathy in the pediatric population.
      In recent years, thanks to the continuous improvement of genetic technologies, several rare variants in different genes, mainly encoding desmosomal proteins, have been identified.
      • Odak M
      • Douedi S
      • Mararenko A
      • et al.
      Arrhythmogenic right ventricular cardiomyopathy: the role of genetics in diagnosis, management, and screening.
      Genetic analysis is often part of clinical diagnosis and it is highly recommended in relatives of an ACM diagnosed family member.
      • Graziano F
      • Zorzi A
      • Cipriani A
      • et al.
      The 2020 “Padua Criteria” for diagnosis and phenotype characterization of arrhythmogenic cardiomyopathy in clinical practice.
      The main gene responsible for almost 40% of ACM cases is the PKP2 gene, encoding the plakophilin-2 protein.
      • Wilde AAM
      • Semsarian C
      • Marquez MF
      • et al.
      European Heart Rhythm Association (EHRA)/Heart Rhythm Society (HRS)/Asia Pacific Heart Rhythm Society (APHRS)/Latin American Heart Rhythm Society (LAHRS) expert consensus statement on the state of genetic testing for cardiac diseases.
      Genetic studies of other components of the cardiac desmosome have been associated with nearly 20% of ACM patients, specifically those genes encoding: desmocollin-2 (DSC2), desmoplakin (DSP), desmoglein-2 (DSG2), and plakoglobin (JUP).
      • Reichart D
      • Lindberg EL
      • Maatz H
      • et al.
      Pathogenic variants damage cell composition and single cell transcription in cardiomyopathies.
      In addition, other minority genes have been also reported as a potential cause of ACM, such as alpha-actinin-2 (ACTN2), cadherin 2 (CDH2), T-catenin (CTNNA3), desmin (DES), filamin C (FLNC), LIM domain binding protein 20 (LDB3), lamin A/C (LMNA), phospholamban (PLN), RNA binding motif protein 20 (RBM20), sodium voltage-gated channel alpha subunit 5 (SCN5A), transforming growth factor-β3 (TFGB3), tight junction protein 1 (TJP1), transmembrane protein 43 (TMEM43), and titin (TTN).
      • de Brouwer R
      • Bosman LP
      • Gripenstedt S
      • et al.
      Value of genetic testing in the diagnosis and risk stratification of arrhythmogenic right ventricular cardiomyopathy.
      Although ACM usually follows an autosomal dominant pattern of inheritance, recessive pattern phenotypes have also been reported in familial Carvajal syndrome (deleterious variant in DSP),
      • Carvajal-Huerta L.
      Epidermolytic palmoplantar keratoderma with woolly hair and dilated cardiomyopathy.
      • Protonotarios N
      • Tsatsopoulou A.
      Naxos disease and Carvajal syndrome: cardiocutaneous disorders that highlight the pathogenesis and broaden the spectrum of arrhythmogenic right ventricular cardiomyopathy.
      Naxos disease (deleterious variant in plakoglobin -JUP-) and deleterious homozygous variants in DSC2
      • Simpson MA
      • Mansour S
      • Ahnood D
      • et al.
      Homozygous mutation of desmocollin-2 in arrhythmogenic right ventricular cardiomyopathy with mild palmoplantar keratoderma and woolly hair.
      (Table 1).
      Table 1Genes and pattern of inheritance associated with ACM
      GeneProteinInheritanceCell structureAnatomic affection
      ACTN2Alpha-actinin-2ADCytoskeletonLV
      CDH2Cadherin 2ADCytoskeletonRV/BiV
      CTNNA3T-cateninADCytoskeletonRV/BiV
      DESDesminADCytoskeletonLV/BiV
      DSC2Desmocollin-2AD / ARDesmosomeRV/BiV
      DSG2Desmoglein-2ADDesmosomeRV/LV/BiV
      DSPDesmoplakinAD / AR (Carvajal)DesmosomeLV/BiV
      FLNCFilamin CADCytoskeletonLV
      JUPPlakoglobinAD / AR (Naxos)CytoskeletonRV/BiV
      LDB3LIM domain binding protein 20ADCytoskeletonRV
      LMNALamin A/CADCytoskeletonLV/BiV
      PLNPhospholambanADCalcium homeostasisLV/BiV
      PKP2Plakophilin-2ADDesmosomeRV/BiV
      RBM20RNA binding motif protein 20ADCytoskeletonLV
      SCN5ASodium voltage-gated channel alpha subunit 5ADSodium transportRV/LV/BiV
      TGFB3Transforming growth factor-β3ADCytoskeletonRV
      TJP1Tight junction proteinADCytoskeletonRV/BiV
      TMEM43Transmembrane protein 43ADCytoskeletonRV/BiV
      TTNTitinADCytoskeletonRV/LV/BiV
      Abbreviations: AD, autosomic dominant; AR, autosomic recessive; BiV, biventricular involvement; LV, left ventricular involvement; RV, right ventricular involvement.
      Nowadays, a comprehensive genetic analysis of ACM-associated genes may unveil the genetic cause in almost 70% of families.
      • Wilde AAM
      • Semsarian C
      • Marquez MF
      • et al.
      European Heart Rhythm Association (EHRA)/Heart Rhythm Society (HRS)/Asia Pacific Heart Rhythm Society (APHRS)/Latin American Heart Rhythm Society (LAHRS) expert consensus statement on the state of genetic testing for cardiac diseases.
      Despite improvement in genetic sequencing, a large part of identified rare variants remains without a conclusive deleterious role in ACM, mainly due to a lack of functional studies.
      • Bourfiss M
      • van Vugt M
      • Alasiri AI
      • et al.
      Prevalence and disease expression of pathogenic and likely pathogenic variants associated with inherited cardiomyopathies in the general population.
      Therefore, a reanalysis is necessary, despite no time frame having been established so far. In order to determine the periodicity of the reanalysis in a previous study by our group, we suggested 5 years as the maximum time for reanalysis of rare variants, mainly those classified with inconclusive/unknown significance (known as VUS) should be performed before clinical translation of genetic results.
      • Vallverdu-Prats M
      • Alcalde M
      • Sarquella-Brugada G
      • et al.
      Rare variants associated with arrhythmogenic cardiomyopathy: reclassification five years later.
      This is a key point, due to the fact that the diagnosis of ACM can be challenging and current guidelines consider genetic data for differential diagnosis. Thus, the identification of a definitely deleterious rare variant may modify the final diagnosis, even though no specific clinical treatment targeting the mutation or the causative gene of the pathology is known yet.

      PATHOPHYSIOLOGICAL MECHANISMS

      The molecular mechanisms by which desmosome deleterious alterations lead to pathological hallmarks of ACM, such as fibrosis, cardiomyocyte loss, adipogenesis, inflammation, and arrhythmogenesis are still poorly defined to date. Probable pathophysiological mechanisms include loss of mechanical integrity at cell-cell junctions, altered signaling pathways at intercalated discs, disruption of ion channels and gap junctions, and aberrant protein trafficking (Fig 1). In some of them, miRNA have already described to be involved.
      Fig 1
      Fig 1Schematic representation of basic pathophysiological mechanisms involved in ACM. Circles indicate triggers and/or other factors involved: blue, genetic mutations; yellow, physical activity; green, androgens; purple, mitochondrial dysfunction and red, miRNA. *Indicates molecular pathways associated to altered miRNA. (For interpretation of the references to color in this figure legend, the reader is referred to the Web version of this article.)

      Cardiomyocyte loss

      Loss of cardiomyocytes is an important feature in the ACM progression. Often, patients present alterations in desmosomal proteins and, therefore, these cell junctions are weak and may have less mechanical strength. This might provoke cell injuries caused by heart contraction stress, resulting in cellular death.
      • Austin KM
      • Trembley MA
      • Chandler SF
      • et al.
      Molecular mechanisms of arrhythmogenic cardiomyopathy.
      This damage may be aggravated by increasing the mechanical stress in the heart during exercise. It is known that in normal conditions, physical activity promotes an upregulation of JUP and JUP-associated proteins, as well as gap junction (GJ) proteins to face the mechanical stress.
      • Kirchhof P
      • Fabritz L
      • Zwiener M
      • et al.
      Age- and training-dependent development of arrhythmogenic right ventricular cardiomyopathy in heterozygous plakoglobin-deficient mice.
      Hence, cardiomyocytes with desmosomal pathogenic alterations are incapable of responding to the stretch pressure caused by a higher level of physical activity, thus altering their mechanical properties and leading to myocytes death.
      • Hariharan V
      • Asimaki A
      • Michaelson JE
      • et al.
      Arrhythmogenic right ventricular cardiomyopathy mutations alter shear response without changes in cell-cell adhesion.
      Another mechanism contributing to cardiomyocyte loss is cardiomyocyte apoptosis; aberrant nuclear localization of plakoglobin in cardiomyocytes acts as an antagonist to Wnt-ß-catenin signaling, leading to apoptosis in an animal model of ACM.
      • Garcia-Gras E
      • Lombardi R
      • Giocondo MJ
      • et al.
      Suppression of canonical Wnt/beta-catenin signaling by nuclear plakoglobin recapitulates phenotype of arrhythmogenic right ventricular cardiomyopathy.
      Moreover, the increase of glycogen synthase kinase 3β (GSK-3β) also contributes to apoptosis by promoting catenin-β1 degradation and impairing the Wnt/ß-catenin pathway.
      • Asimaki A
      • Kapoor S
      • Plovie E
      • et al.
      Identification of a new modulator of the intercalated disc in a zebrafish model of arrhythmogenic cardiomyopathy.
      Finally, activation of the EP300-TP53 pathway, known to suppress the Hippo and canonical Wnt pathways, was detected in right and left ventricular endomyocardial biopsy from ACM patients, and was associated with increased apoptosis, suggesting a pathogenic role during ACM.
      • Rouhi L
      • Fan S
      • Cheedipudi SM
      • et al.
      The EP300/TP53 pathway, a suppressor of the Hippo and canonical WNT pathways, is activated in human hearts with arrhythmogenic cardiomyopathy in the absence of overt heart failure.

      Fibrosis

      Cardiac fibrosis is a common pathological change associated with cardiac injuries and disease. Since the heart is not able to regenerate its muscular tissue, cardiomyocytes are substituted by cardiac fibroblasts, which proliferate and differentiate in myofibroblasts, resulting in excessive extracellular matrix (ECM) deposition and consequent mechanical stiffness.
      • Banerjee I
      • Yekkala K
      • Borg TK
      • Baudino TA.
      Dynamic interactions between myocytes, fibroblasts, and extracellular matrix.
      In ACM patients, fibrosis is promoted by complex interactions between cytokines, growth factors and hormones.
      • Austin KM
      • Trembley MA
      • Chandler SF
      • et al.
      Molecular mechanisms of arrhythmogenic cardiomyopathy.
      The transforming growth factor beta (TGF-β) is a pivotal cytokine/growth factor that contributes to cardiac fibrosis by phosphorylating and activating the receptor-associated SMAD, that triggers the expression of profibrotic genes such as ECM proteins and inhibitors of matrix metalloproteinases (MMP).
      • Dobaczewski M
      • Gonzalez-Quesada C
      • Frangogiannis NG.
      The extracellular matrix as a modulator of the inflammatory and reparative response following myocardial infarction.
      Li et al
      • Li CL
      • Leng JH
      • Li MH
      • Shi JH
      • Jia SZ
      • Lang JH.
      Expressions and roles of TGFbeta/Smad signal pathway in peritoneum of endometriosis.
      showed increased TGFβ1 expression levels and activation of SMAD2 signaling in an ACM mouse model associated with a rare variant in the JUP gene. Moreover, the β-adrenergic system could also regulate the extracellular matrix protein turnover. Norepinephrine could trigger the expression of tissue inhibitor of metalloproteinases 1 (TIMP-1), by decreasing metalloproteinase 2 MMP-2/TIMP-1 ratio.
      • Maione AS
      • Pilato CA
      • Casella M
      • et al.
      Fibrosis in arrhythmogenic cardiomyopathy: the phantom thread in the fibro-adipose tissue.
      ,
      • Meier H
      • Bullinger J
      • Deten A
      • et al.
      Tissue inhibitor of matrix metalloproteinase-1 in norepinephrine-induced remodeling of the mouse heart.
      Mechanical stress caused by a high level of physical activity could increase plasmatic levels of TGFβ1.
      • Maione AS
      • Pilato CA
      • Casella M
      • et al.
      Fibrosis in arrhythmogenic cardiomyopathy: the phantom thread in the fibro-adipose tissue.
      On the other hand, the noncanonical TGFβ pathway also plays a role in cardiac fibrosis by activation of mitogen-activated protein kinase (MAPK) signaling and generation of reactive oxygen species.
      • Jain M
      • Rivera S
      • Monclus EA
      • et al.
      Mitochondrial reactive oxygen species regulate transforming growth factor-beta signaling.
      A knockdown of pkp2 in a cell model presented an upregulation of fibrotic genes via noncanonical TGFβ–MAPK signalling.
      • Dubash AD
      • Kam CY
      • Aguado BA
      • et al.
      Plakophilin-2 loss promotes TGF-beta1/p38 MAPK-dependent fibrotic gene expression in cardiomyocytes.
      All these data suggest that both canonical and noncanonical TGFβ signaling can have a role in inducing myocardial fibrosis in ACM. Fibrosis is magnified by a positive loop promoted by changes in extracellular matrix composition that vary tissue properties by increasing its rigidity. Subsequently, tissue stiffness promotes the differentiation of myofibroblasts that produce and release collagen, thus further increasing the stiffness of the tissue.
      • Maione AS
      • Pilato CA
      • Casella M
      • et al.
      Fibrosis in arrhythmogenic cardiomyopathy: the phantom thread in the fibro-adipose tissue.

      Inflammation

      Inflammatory infiltrates are frequently observed in ACM biopsy samples, commonly in fibrotic areas.
      • Thiene G
      • Corrado D
      • Nava A
      • et al.
      Right ventricular cardiomyopathy: is there evidence of an inflammatory aetiology?.
      • Basso C
      • Thiene G
      • Corrado D
      • Angelini A
      • Nava A
      • Valente M.
      Arrhythmogenic right ventricular cardiomyopathy. Dysplasia, dystrophy, or myocarditis?.
      • Corrado D
      • Basso C
      • Thiene G
      • et al.
      Spectrum of clinicopathologic manifestations of arrhythmogenic right ventricular cardiomyopathy/dysplasia: a multicenter study.
      Myocarditis, mainly caused by cardiotropic viruses’ infection, may herald the onset of ACM and contributes to its progression.
      • Calabrese F
      • Basso C
      • Carturan E
      • Valente M
      • Thiene G.
      Arrhythmogenic right ventricular cardiomyopathy/dysplasia: is there a role for viruses?.
      Differential diagnosis between myocarditis and ACM is still challenging and difficult to distinguish in some patients who show an overlap of clinical features, such as desmosome disruption, inflammatory infiltrates and the presence of viral genomic material.
      • Campuzano O
      • Alcalde M
      • Iglesias A
      • et al.
      Arrhythmogenic right ventricular cardiomyopathy: severe structural alterations are associated with inflammation.
      Some authors suggest that ACM could be a form of myocarditis due to this clinical overlap.
      • Basso C
      • Thiene G
      • Corrado D
      • Angelini A
      • Nava A
      • Valente M.
      Arrhythmogenic right ventricular cardiomyopathy. Dysplasia, dystrophy, or myocarditis?.
      Moreover, this hypothesis has been published as a case report that presents a patient who died of myocarditis but also fit in ACM diagnosis. Recently, some studies state that nearly 10% of cases diagnosed with myocarditis carried deleterious variants in genes associated with ACM.
      • Lota AS
      • Hazebroek MR
      • Theotokis P
      • et al.
      Genetic architecture of acute myocarditis and the overlap with inherited cardiomyopathy.
      • Seidel F
      • Laser KT
      • Klingel K
      • et al.
      Pathogenic variants in cardiomyopathy disorder genes underlie pediatric myocarditis-further impact of heterozygous immune disorder gene variants?.
      • Kissopoulou A
      • Fernlund E
      • Holmgren C
      • et al.
      Monozygotic twins with myocarditis and a novel likely pathogenic desmoplakin gene variant.
      • Tiron C
      • Campuzano O
      • Fernandez-Falgueras A
      • et al.
      Prevalence of pathogenic variants in cardiomyopathy-associated genes in myocarditis.
      • Campuzano O
      • Fernandez-Falgueras A
      • Sarquella-Brugada G
      • et al.
      A genetically vulnerable myocardium may predispose to myocarditis.
      It has been suggested that desmosomal dysfunction stimulates the production of inflammatory molecules, but it is not clear whether myocardial inflammation is a primary cause of ACM or just a secondary response to cardiomyocyte death.
      • Meraviglia V
      • Alcalde M
      • Campuzano O
      • Bellin M.
      Inflammation in the pathogenesis of arrhythmogenic cardiomyopathy: secondary event or active driver?.

      Adipogenesis

      Adipogenesis is a common feature of ACM that consists of the infiltration of adipocytes, in this case, into the heart tissue. The cellular source of these adipocytes and the molecular signals are still under discussion. Regarding the cellular origin of adipocytes, there are several candidates: cardiomyocytes, differentiated nonmyocytes, cardiac progenitor cells and circulating progenitor cells.
      • Kohela A
      • van Rooij E.
      Fibro-fatty remodelling in arrhythmogenic cardiomyopathy.
      There are numerous studies in mouse models showing that second heart field progenitor cells are involved in the origin of adipocytes.
      • Lombardi R
      • Dong J
      • Rodriguez G
      • et al.
      Genetic fate mapping identifies second heart field progenitor cells as a source of adipocytes in arrhythmogenic right ventricular cardiomyopathy.
      • Moses KA
      • DeMayo F
      • Braun RM
      • Reecy JL
      • Schwartz RJ.
      Embryonic expression of an Nkx2-5/Cre gene using ROSA26 reporter mice.
      • Verzi MP
      • McCulley DJ
      • De Val S
      • Dodou E
      • Black BL.
      The right ventricle, outflow tract, and ventricular septum comprise a restricted expression domain within the secondary/anterior heart field.
      • Zhou B
      • von Gise A
      • Ma Q
      • Rivera-Feliciano J
      • Pu WT.
      Nkx2-5- and Isl1-expressing cardiac progenitors contribute to proepicardium.
      Moreover, another study proved the ability of cardiomyocytes to transdifferentiate into adipocytes in vitro.
      • Dorn T
      • Kornherr J
      • Parrotta EI
      • et al.
      Interplay of cell-cell contacts and RhoA/MRTF-A signaling regulates cardiomyocyte identity.
      All these data contribute to elucidate the origin of adipocytes, but more studies are needed to reach a consensus about their source in ACM. On the other hand, molecular signals triggering adipogenesis are also under investigation. Several signaling pathways have been described to be involved in adipogenesis.
      • Austin KM
      • Trembley MA
      • Chandler SF
      • et al.
      Molecular mechanisms of arrhythmogenic cardiomyopathy.
      There is evidence that the downregulation of the Wnt pathway contributes to adipogenesis by triggering the differentiation of mesenchymal stem cells and preadipocytes into adipocytes.
      • Laudes M.
      Role of WNT signalling in the determination of human mesenchymal stem cells into preadipocytes.
      In the absence of Wnt ligands, GSK-3β phosphorylates catenin-β1. The interaction between GSK-3β and catenin-β1 is stabilized by Axin, which serves as a negative regulator of the canonical Wnt pathway.
      • Gao S
      • Puthenvedu D
      • Lombardi R
      • Chen SN.
      Established and emerging mechanisms in the pathogenesis of arrhythmogenic cardiomyopathy: a multifaceted disease.
      ,
      • Willert K
      • Shibamoto S
      • Nusse R.
      Wnt-induced dephosphorylation of axin releases beta-catenin from the axin complex.
      Also, in the presence of Wnt ligands, GSK-3β no longer phosphorylates catenin-β1, that accumulates in the cytoplasm and translocates into the nucleus where it co-activates the T-cell/lymphoid enhancer (TCF-LEF) transcription factors that will upregulate the expression of adipogenic genes.
      • Chen S
      • Guttridge DC
      • You Z
      • et al.
      Wnt-1 signaling inhibits apoptosis by activating beta-catenin/T cell factor-mediated transcription.
      • Hoorntje ET
      • Te Rijdt WP
      • James CA
      • et al.
      Arrhythmogenic cardiomyopathy: pathology, genetics, and concepts in pathogenesis.
      • Niehrs C.
      The complex world of WNT receptor signalling.

      BIOMARKERS

      Early diagnosis and risk stratification are crucial for identifying at-risk patients who need therapeutic intervention. The features of a biomarker include high specificity, accuracy, reproducibility, ease procurement and a low price. A biological marker needs to add value over the existing diagnostic tests, being useful for clinical decision-making. One of the main limitations of biomarkers related to ACM is their lack of specificity, not being useful in optimizing clinical decision-making in this population. Some of the proposed biomarkers, such as troponin I (cTNI) or heat shock protein 70 (HSP70), have not shown any disease-specificity. Concretely, cTNI has been associated with ACM, but this protein informs us more about a myocardial injury than disease-specificity, not leading to a certain CVD etiology. Thus, apoptosis and impairment of the intercalated discs may justify a release of this myocardial protein in ACM.
      • Kostis WJ
      • Tedford RJ
      • Miller DL
      • Schulman SP
      • Tomaselli GF.
      Troponin-I elevation in a young man with arrhythmogenic right ventricular dysplasia/cardiomyopathy.
      However, cTNI plasmatic concentration may be increased in several situations, such as extreme exercise, myocarditis or myocardial ischemia, among others. HSP70 is a chaperone which was described to be associated with damaged myocardial cells in ACM failing hearts compared to nonfailing hearts
      • Wei YJ
      • Huang YX
      • Shen Y
      • et al.
      Proteomic analysis reveals significant elevation of heat shock protein 70 in patients with chronic heart failure due to arrhythmogenic right ventricular cardiomyopathy.
      ; however, a high level of this protein has also been found in dilated and ischemic cardiomyopathies in response to tissue damage. Bridging integrator 1 (BIN1) is a protein that belongs to the BAR domain superfamily and organizes membrane structure and function. BIN1 is involved in cardiomyocyte calcium homeostasis and, when altered, leads to an excitation-contraction impairment. Hong et al
      • Hong TT
      • Cogswell R
      • James CA
      • et al.
      Plasma BIN1 correlates with heart failure and predicts arrhythmia in patients with arrhythmogenic right ventricular cardiomyopathy.
      established a correlation between BIN1 and heart failure in a small cohort of ACM patients. Although BIN1 is not directly linked to cardiac desmosomes, reduced concentrations of this protein predicted ventricular arrhythmia events in this population. Nevertheless, this marker does not discriminate between early-stage ACM patients without heart failure and healthy subjects, furthermore, BIN1 leads to a pathologic cardiomyocyte alteration more than to contractile dysfunction.
      Currently, some of the proposed circulating blood biomarkers remain potential specific markers for late phase ACM. Brain natriuretic peptide (BNP) has been described as an indicator of ventricular dysfunction in ACM patients, with a negative correlation with RV ejection fraction, helping to discriminate between idiopathic ventricular tachycardia (IVT) and ACM.
      • Matsuo K
      • Nishikimi T
      • Yutani C
      • et al.
      Diagnostic value of plasma levels of brain natriuretic peptide in arrhythmogenic right ventricular dysplasia.
      Recently, N-terminal fragment of BNP (NT-pro-BNP) was positively correlated with RV end-diastolic and end-systolic diameter, as well as negatively associated with RV ejection fraction assessed by cardiac magnetic resonance.
      • Bueno Marinas M
      • Celeghin R
      • Cason M
      • et al.
      A microRNA expression profile as non-invasive biomarker in a large arrhythmogenic cardiomyopathy cohort.
      ,
      • Cheng H
      • Lu M
      • Hou C
      • et al.
      Relation between N-terminal pro-brain natriuretic peptide and cardiac remodeling and function assessed by cardiovascular magnetic resonance imaging in patients with arrhythmogenic right ventricular cardiomyopathy.
      Moreover, NT-proBNP confirmed to be increased in patients with biventricular ACM.
      • Akdis D
      • Saguner AM
      • Burri H
      • et al.
      Clinical predictors of left ventricular involvement in arrhythmogenic right ventricular cardiomyopathy.
      The main flaw of these molecules is their low specificity.
      • Stadiotti I
      • Pompilio G
      • Maione AS
      • Pilato CA
      • D'Alessandra Y
      • Sommariva E
      Arrhythmogenic cardiomyopathy: what blood can reveal?.
      Another circulating marker is interleukin-33 receptor ST2, associated with fibrosis and cardiac remodeling, which has been correlated to RV and/or left ventricular dysfunction and potential lethal arrhythmias in the ACM population.
      • Holte E
      • Kleveland O
      • Ueland T
      • et al.
      Effect of interleukin-6 inhibition on coronary microvascular and endothelial function in myocardial infarction.
      Finally, Galectin-3 (GAL-3), an inflammatory protein related to fibrosis, was also found to be increased in plasma of ACM patients, thus correlating with more susceptibility to ventricular arrhythmia.
      • Oz F
      • Onur I
      • Elitok A
      • et al.
      Galectin-3 correlates with arrhythmogenic right ventricular cardiomyopathy and predicts the risk of ventricular-arrhythmias in patients with implantable defibrillators.
      This marker has also been demonstrated to be elevated in heart failure cohorts at risk of sudden death due to VF or VT.
      • Francia P
      • Adduci C
      • Semprini L
      • et al.
      Osteopontin and galectin-3 predict the risk of ventricular tachycardia and fibrillation in heart failure patients with implantable defibrillators.
      Several studies have also identified increased levels of pro-inflammatory cytokines in the plasma and serum of ACM patients, thus suggesting them as candidate circulating biomarkers.
      • Campian ME
      • Verberne HJ
      • Hardziyenka M
      • et al.
      Assessment of inflammation in patients with arrhythmogenic right ventricular cardiomyopathy/dysplasia.
      ,
      • Asimaki A
      • Tandri H
      • Duffy ER
      • et al.
      Altered desmosomal proteins in granulomatous myocarditis and potential pathogenic links to arrhythmogenic right ventricular cardiomyopathy.
      Likewise, C‑reactive protein was detected at higher levels in the plasma of ACM patients immediately after an episode of ventricular arrhythmia, compared to patients with IVT, thus suggesting a correlation with arrhythmic events in ACM, unrelated to structural and/or functional anomalies of the heart.
      • Bonny A
      • Lellouche N
      • Ditah I
      • et al.
      C-reactive protein in arrhythmogenic right ventricular dysplasia/cardiomyopathy and relationship with ventricular tachycardia.
      However, the specificity of these circulating inflammatory mediators remains limited for ACM, since elevated cytokine levels have been described in several cardiovascular diseases.

      NONCODING RNAS

      The diagnosis of ACM is based on a scoring system with major and minor criteria.
      • Marcus FI
      • McKenna WJ
      • Sherrill D
      • et al.
      Diagnosis of arrhythmogenic right ventricular cardiomyopathy/dysplasia: proposed modification of the task force criteria.
      However, an early identification of ACM is particularly difficult, taking into account the absence of a single clinical standard to reach the diagnosis, overlapping phenotypes with other cardiac diseases, incomplete penetrance and variable phenotype expressivity, as well as the genetic diagnosis limitations. Moreover, the lack of specific diagnosis tools, either imaging tests or circulating markers, leads to an urgent need for novel biomarkers in ACM. Steady progress in the discovery of new biomarkers and more sophisticated clinical applications offer promising possibilities for enhancing the care of patients.
      In the last few years, researchers and clinicians have identified noncoding RNAs (ncRNAs) as useful biomarkers due to their presence in biological fluids, such as plasma and serum. ncRNA have a wide variety of types based on function and size (Fig 2, Table 2). Many cardiovascular diseases have shown an aberrant expression of ncRNAs in cardiac cells involved in pivotal regulatory roles in pathophysiological processes, and several reports have described the diagnostic potential of circulating ncRNAs in cardiovascular diseases.
      • Gabriel AF
      • Costa MC
      • Enguita FJ.
      Interactions among regulatory non-coding RNAs involved in cardiovascular diseases.
      ,
      • Jiang W
      • Agrawal DK
      • Boosani CS.
      Non-coding RNAs as epigenetic gene regulators in cardiovascular diseases.
      Specific patterns of ncRNA expression have been related to certain cardiovascular disorders.
      • van Rooij E
      • Sutherland LB
      • Liu N
      • et al.
      A signature pattern of stress-responsive microRNAs that can evoke cardiac hypertrophy and heart failure.
      These molecules are of great value as diagnostic/prognostic biomarkers for heart failure and several cardiac diseases. Thus, ncRNAs are a group of genes without protein codification. Briefly, they can be divided into housekeeping ncRNAs, mainly tRNAs, rRNAs, and snRNAs, which play crucial roles in cellular processes. Others are regulatory ncRNAs, mainly miRNAs, small interfering RNAs (siRNAs) and long noncoding RNAs (lncRNAs), which are tissue-specific and work in response to various internal and environmental stimuli.
      Fig 2
      Fig 2Classification of noncoding RNA. Noncoding RNA can be divided into 2 groups: housekeeping and regulatory ncRNA. The regulatory ncRNA can also be divided into 2 groups based on their size (long and short ncRNA). (For interpretation of the references to color in this figure legend, the reader is referred to the Web version of this article.)
      Table 2Main noncoding RNA (ncRNA) classes
      NameAcronymLength (nt)Description
      Circular RNAcircRNA≈100–1600Covalently closed RNA rings; some have coding functions; potential gene regulators.
      Small interferingsiRNA20–25Double-stranded RNAs similar to miR, operating through RNA interference (RNAi) pathway; promote mRNA degradation
      Micro-RNAmiRNA/miR21–24Function in RNA silencing and post-transcriptional regulation of gene expression; may have an extracellular localization
      Piwi-interacting RNApiRNA26–31Epigenetic and post-transcriptional gene silencing of retrotransposons and other genetic elements in germ line cells.
      Small nuclear RNAssnoRNAs≈60–200Guide chemical modifications of other RNAs (rRNA, tRNA, snRNA)
      Small nuclear RNAssnRNA/U-RNA≈150Function in the processing of pre-messenger RNA (hnRNA) in the nucleus; aid in the regulation of transcription factors; telomere maintenance
      Natural antisense transcriptsNAT>200RNAs encoded within a cell that have transcript complementarity to other RNA transcripts; RNA interference (RNAi); alternative splicing, genomic imprinting; X-chromosome inactivation.
      Enhancer RNAseRNA50–2000Transcriptional regulation in cis and in trans
      Long noncoding RNAs are considered >200 nt in length. Short noncoding RNAs are considered <200 nt.
      MiRNAs have emerged as main players in human physiological and pathologic processes, including cardiovascular phenotypes.
      • Calderon-Dominguez M
      • Mangas A
      • Belmonte T
      • Quezada-Feijoo M
      • Ramos M
      • Toro R.
      Ischemic dilated cardiomyopathy pathophysiology through microRNA-16-5p.
      In recent years, the study of miRNAs as potential biomarkers in ACM has progressively increased. This review provides an overview on the role of microRNAs (miRNAs) in ACM, focusing on the modulated biological pathways and their potential use as potential biomarkers; moreover, we discuss future clinical applications and limitations of this novel tool.

      MicroRNAs

      MiRNAs are single-stranded, small RNA comprising 19–25 nucleotides endogenously expressed, evolutionarily conserved in animals.
      • Small EM
      • Olson EN.
      Pervasive roles of microRNAs in cardiovascular biology.
      MiRNAs are involved in complex networks that allow interaction and control of the cellular phenotype. Most of the biological processes such as cell cycle control, proliferation, differentiation, inflammation, apoptosis, autophagy, or stress response are affected by miRNAs.
      • Beermann J
      • Piccoli MT
      • Viereck J
      • Thum T.
      Non-coding RNAs in development and disease: background, mechanisms, and therapeutic approaches.
      Their principal function is the regulation of gene expression through translational inhibition and/or promoting degradation of target messenger RNA (mRNA). They show a pleiotropic function as a single miRNA that can regulate different mRNAs, and each of the regulated protein-coding genes can be modulated by various miRNAs, thus allowing cooperation between miRNAs. Also, miRNAs themselves can be modulated by feedback mechanisms promoted by the protein products of mRNA targets.
      • Liu N
      • Olson EN.
      MicroRNA regulatory networks in cardiovascular development.
      The general mechanism of miRNA action is based on pairing the so called ‘‘seed’’ sequence (miRNA nt 2–8) with complementary sites of the target messenger RNA, mainly located in the 3′-untranslated region (3′UTR), but also in the coding region or in the 5′UTR The role of RNA structure at 5′ untranslated region in microRNA-mediated gene regulation.
      • Gu W
      • Xu Y
      • Xie X
      • Wang T
      • Ko JH
      • Zhou T.
      The role of RNA structure at 5′ untranslated region in microRNA-mediated gene regulation.
      ,
      • Brummer A
      • Hausser J.
      MicroRNA binding sites in the coding region of mRNAs: extending the repertoire of post-transcriptional gene regulation.
      A full or partial miRNA complementarity with the targeted mRNAs can determine the nature of target repression.
      • Bartel DP.
      MicroRNAs: target recognition and regulatory functions.
      Complete base-pairing leads to mRNA endonucleolytic cleavage, while partial base-pairing results in the repression of mRNA translation through interference with the translational machinery (directed to polyribosomes in subcellular compartments, resulting in truncated protein) or by sequestering in cytoplasmic P-bodies. To date, over 2500 human mature miRNAs have been annotated in the miRBase (http://www. mirbase.org/).

      BIOGENESIS OF MICRORNAS

      The biogenesis of miRNAs consists of several steps from the nucleus to the cytoplasm. MiRNAs can be transcribed individually or in clusters. Based on their locations in the genome, miRNAs are classified as intergenic with their own promoter, or exonic and intronic. Their endonuclease synthesis is mediated through a Pol II transcription, resulting in a precursor molecule, called primary miRNA transcript (pri-miRNA) that shows a distinctive hairpin structure. Pri-miRNA is processed by a nuclear complex, comprising the DGCR8 protein (DiGeorge syndrome critical region 8) that recognizes the pri-miRNA and directs the cleavage by RNAse III endonuclease Drosha. The pri-miRNA processed by Drosha/GCR8 is converted into 60–100 nt hairpins, called precursor miRNAs (pre-RNAs), exported from the nucleus to the cytoplasm by Exportin 5/Ran-GTP action. Once there, pre-miRNA is cleaved by ribonuclease III enzyme Dicer/TRBP and turns out into a miRNA duplex of about 20 bp. Finally, the miRNA duplex merge with a protein named argonaute-2 (AGO2) and is incorporated into RNA-induced silencing complex (RISC), in which one strand is degraded, while the other constitutes the mature miRNA. Then, the miRNA-RISC complex recognizes specific targets, acting as regulators of gene expression and inducing gene silencing at posttranscriptional level.
      • Hayder H
      • O'Brien J
      • Nadeem U
      • Peng C.
      MicroRNAs: crucial regulators of placental development.
      Notably, miRNA biogenesis can follow an alternative noncanonical nuclear pathway through the generation of precursors derived from short hairpin introns, named “mirtrons.”
      • Okamura K
      • Hagen JW
      • Duan H
      • Tyler DM
      • Lai EC.
      The mirtron pathway generates microRNA-class regulatory RNAs in Drosophila.

      FINDINGS ON MIRNAS (INVOLVEMENT) IN ACM

      Circulating miRNAs in human samples

      The current interest in miRNAs as biomarkers is justified by their capacity to circulate extracellularly in biofluids, within membranous vesicles or associated with RNA-binding proteins and high-and low-density lipoprotein particles,
      • Freedman JE
      • Gerstein M
      • Mick E
      • et al.
      Diverse human extracellular RNAs are widely detected in human plasma.
      miRNAs integrity in the extracellular environment. During the last few years, several circulating miRNAs have been associated with ACM. Sommariva et al
      • Sommariva E
      • D'Alessandra Y
      • Farina FM
      • et al.
      MiR-320a as a potential novel circulating biomarker of arrhythmogenic cardiomyopathy.
      performed a profile of circulating miRNAs in 2017 by evaluating the plasmatic expression of 377 miRNAs in ACM patients and healthy individuals, and then validating their results in 36 ACM, 21 IVT patients and 53 healthy controls. Only miR-320a showed statistically significant differences with 0.53- and 0.78-fold expression differences in ACM vs IVT patients and healthy cohorts, respectively. Briefly, these results suggested that miR-320a could be a novel potential biomarker for the disease, to distinguish between ACM and IVT. Of note, miR-320a was upregulated in human mesenchymal (skeletal or stromal) cells during adipogenic differentiation, thus suggesting mechanistic implications in ACM pathogenesis.
      • Hamam D
      • Ali D
      • Vishnubalaji R
      • et al.
      microRNA-320/RUNX2 axis regulates adipocytic differentiation of human mesenchymal (skeletal) stem cells.
      Similarly, Yamada et al
      • Yamada S
      • Hsiao YW
      • Chang SL
      • et al.
      Circulating microRNAs in arrhythmogenic right ventricular cardiomyopathy with ventricular arrhythmia.
      screened 84 cardiac-related miRNAs in a cohort of 62 patients (28 ACM, 11 borderline or possible ACM, 23 IVT) and 33 healthy patients. Four circulating miRNAs (miR-144-3p, miR-145-5p, miR-185-5p, and miR-494) showed higher expression levels in the ACM cohort compared to the other 3 groups. Interestingly, miR-494 was also a predictive factor of recurrent ventricular arrhythmia after ablation in ACM subjects.
      • Yamada S
      • Hsiao YW
      • Chang SL
      • et al.
      Circulating microRNAs in arrhythmogenic right ventricular cardiomyopathy with ventricular arrhythmia.
      According to these results, the screening of 754 miRNAs in the plasma of 21 ACM patients distributed based on their genotype or phenotype, versus 20 healthy individuals showed the overexpression of miR185-5p in the ACM plasma group.
      One of the most solid studies on ACM and miRNAs screened miRNAs on right ventricular tissue and blood samples of 9 ACM patients and 4 controls. Afterward, a unique set of 6 circulating miRNAs was validated on 90 total blood samples from an independent cohort. This miRNA set showed high discriminatory diagnostic power in ACM patients when compared to controls, nonaffected family members of ACM probands carrying a desmosomal pathogenic variant, and other cardiomyopathy groups such as hypertrophic cardiomyopathy, dilated cardiomyopathy and myocarditis. Concretely, miR-122-5p, miR-182-5p, and miR-183-5p were overexpressed in ACM compared to controls and the other cardiomyopathies, while miR-133a-3p, miR-133b, and miR-142-3p were downregulated in ACM compared to controls and other groups.
      • Bueno Marinas M
      • Celeghin R
      • Cason M
      • et al.
      A microRNA expression profile as non-invasive biomarker in a large arrhythmogenic cardiomyopathy cohort.
      Only the ACM cohort fulfilled the miRNA set evolving as a potential biomarker for the clinical practice.

      Cardiac miRNAs in human heart tissues

      In 2007, the gap protein Connexin-43 (encoded by the GJA1 gene) was involved with malignant arrhythmogenic events. Bioinformatics analysis demonstrated that miR-130 targets connexin-43, and it is up regulated in cardiomyopathies with heart failure.
      • Thum T
      • Galuppo P
      • Wolf C
      • et al.
      MicroRNAs in the human heart: a clue to fetal gene reprogramming in heart failure.
      In 2016, Zhang et al
      • Zhang H
      • Liu S
      • Dong T
      • et al.
      Profiling of differentially expressed microRNAs in arrhythmogenic right ventricular cardiomyopathy.
      performed the first expression profile of miRNAs on a cohort of 24 ACM patients who underwent cardiac transplants, compared to a group of 24 healthy controls. This study showed a set of 21 validated miRNAs as potential ACM miRNA signature, where 11 were significantly upregulated miRNAs (miR-21-3p, miR-21-5p, miR34a-5p, miR-212-3p, miR-216a, miR-584-3p, miR-1251, miR-3621, miR-3692-3p, miR-4286, miR-4301), and 10 were downregulated miRNAs (miR-135b, miR-138-5p, miR-193b-3p, miR-302b-3p, miR-302c-3p, miR-338-3p, miR-491-3p, miR-575, miR-4254, miR-4643) in an ACM heart, compared to a healthy control. Importantly, functional enrichment analysis established a correlation between the target genes of miR-21-5p and miR-135b and Wnt and Hippo signaling pathways, molecular mechanisms related to the ACM pathophysiology, suggesting a possible role in fibro-fatty substitution.
      • Bueno Marinas M
      • Celeghin R
      • Cason M
      • et al.
      A microRNA expression profile as non-invasive biomarker in a large arrhythmogenic cardiomyopathy cohort.
      Although in the relevance of this study, a validation cohort was lacking and the arbitrary selection of miR21-5p and miR-135b over others with higher differential expression was not entirely justified (Table 3).
      Table 3The main research based on ACM and miRNAs
      Patients/modelSourcePathogenic alterationMain findingsReference
      Screening of 377 miRNAs on ACM and HC subjects Validation on 36 ACM,
      • Seidel F
      • Laser KT
      • Klingel K
      • et al.
      Pathogenic variants in cardiomyopathy disorder genes underlie pediatric myocarditis-further impact of heterozygous immune disorder gene variants?.
      HC,
      • Wei YJ
      • Huang YX
      • Shen Y
      • et al.
      Proteomic analysis reveals significant elevation of heat shock protein 70 in patients with chronic heart failure due to arrhythmogenic right ventricular cardiomyopathy.
      IVT
      • Asimaki A
      • Kapoor S
      • Plovie E
      • et al.
      Identification of a new modulator of the intercalated disc in a zebrafish model of arrhythmogenic cardiomyopathy.
      individuals
      PlasmamiR-320a was overexpressed in ACM vs IVT. Not correlated with ACM severitySommariva et al
      • Sommariva E
      • D'Alessandra Y
      • Farina FM
      • et al.
      MiR-320a as a potential novel circulating biomarker of arrhythmogenic cardiomyopathy.
      Screening 84 cardiac-related miRNAs expression profiles in ACM
      • Gerull B
      • Brodehl A.
      Insights into genetics and pathophysiology of arrhythmogenic cardiomyopathy.
      vs HC
      • Gerull B
      • Brodehl A.
      Insights into genetics and pathophysiology of arrhythmogenic cardiomyopathy.
      Validation using RT-PCR on definitive ACM,
      • Jain M
      • Rivera S
      • Monclus EA
      • et al.
      Mitochondrial reactive oxygen species regulate transforming growth factor-beta signaling.
      borderline ACM,
      • de Brouwer R
      • Bosman LP
      • Gripenstedt S
      • et al.
      Value of genetic testing in the diagnosis and risk stratification of arrhythmogenic right ventricular cardiomyopathy.
      IVT,
      • Banerjee I
      • Yekkala K
      • Borg TK
      • Baudino TA.
      Dynamic interactions between myocytes, fibroblasts, and extracellular matrix.
      HC
      • Calabrese F
      • Basso C
      • Carturan E
      • Valente M
      • Thiene G.
      Arrhythmogenic right ventricular cardiomyopathy/dysplasia: is there a role for viruses?.
      Plasma Screening microarray Validation RT-PCRDefinite ACM patients had significantly higher expression of circulating miR-144-3p, 145-5p, 185-5p, and 494 miR-494 was overexpressed in definitive ACM with recurrent VA after catheter ablationYamada et al
      • Yamada S
      • Hsiao YW
      • Chang SL
      • et al.
      Circulating microRNAs in arrhythmogenic right ventricular cardiomyopathy with ventricular arrhythmia.
      Screening 754 miRNAs in 4 ACM and 4 HC samples.

      Validation on ACM
      • Kissopoulou A
      • Fernlund E
      • Holmgren C
      • et al.
      Monozygotic twins with myocarditis and a novel likely pathogenic desmoplakin gene variant.
      and HC
      • Thiene G
      • Corrado D
      • Nava A
      • et al.
      Right ventricular cardiomyopathy: is there evidence of an inflammatory aetiology?.
      Plasma

      Taqman Array Human micro-RNA
      PKP2, DSP, DSG2miR-185-5p as a potential marker of ACMSacchetto et al
      • Sacchetto C
      • Mohseni Z
      • Colpaert RMW
      • et al.
      Circulating miR-185-5p as a potential biomarker for arrhythmogenic right ventricular cardiomyopathy.
      Screening of miRNAs in 9 ACM and 4 HC using 84 miRNAs array

      Validation on the largest cohort ACM blood samples
      • Osbourne A
      • Calway T
      • Broman M
      • McSharry S
      • Earley J
      • Kim GH.
      Downregulation of connexin43 by microRNA-130a in cardiomyocytes results in cardiac arrhythmias.
      and HC,
      • D'Ascenzi F
      • Valentini F
      • Pistoresi S
      • et al.
      Causes of sudden cardiac death in young athletes and non-athletes: systematic review and meta-analysis: sudden cardiac death in the young.
      nonaffected family members and others cardiomyopathies
      Heart tissue and plasma

      Small RNA sequence in tissue samples 84 miRNAs cardiac-related arrays for plasma by NGS analysis
      miR-122-5p, miR-182-5p, and miR-183-5p were overexpressed in ACMBueno et al
      • Bueno Marinas M
      • Celeghin R
      • Cason M
      • et al.
      A microRNA expression profile as non-invasive biomarker in a large arrhythmogenic cardiomyopathy cohort.
      Screening 1078 miRNAs on 24 HTX and 24 HCMyocardial tissue

      Using S-Poly(T) Plus method
      11 upregulated: miR-21-3p, miR-21-5p, miR34a-5p, miR-212-3p, miR-216a, miR-584-3p, miR-1251, miR-3621, miR-3692-3p, miR-4286, miR-4301

      10 downregulated: miR-135b, miR-138-5p, miR-193b-3p, miR-302b-3p, miR-302c-3p, miR-338-3p, miR-491-3p, miR-575, miR-4254, miR-4643.

      MiR-21-5p and miR-135b known to be involved as modulators of Wnt and Hippo pathways
      Zhang et al
      • Zhang H
      • Liu S
      • Dong T
      • et al.
      Profiling of differentially expressed microRNAs in arrhythmogenic right ventricular cardiomyopathy.
      Screening on 3 ACM patients and 3 HC. For validation, RT-qPCR measurements was performed of miR-520c-3p and miR-29b-3p in 8 AMC and 5 HC, that included those of the screening stepCardiac stromal cells derived from endomyocardial biopsies of ACM patients

      Using TaqMan Low Density Arrays
      miR-29b-3p in cardiac stromal cells contributes to ACM pathogenesisRainer et al
      • Rainer J
      • Meraviglia V
      • Blankenburg H
      • et al.
      The arrhythmogenic cardiomyopathy-specific coding and non-coding transcriptome in human cardiac stromal cells.
      Control and PKP2-knock down HL-1 (HL-1Pkp2-shRNA) cells were screened for 750 miRNAsPKP2-deficient HL-1 cell (HL-1PKP2-shRNA) lines Using low Taqman miRNAarray analysesMiRNA-184, the most down regulated, was predominantly expressed in cardiac mesenchymal progenitor cells

      Knock down of miR-184 in HL-1 cells and MPCs induced and conversely, its over-expression attenuated adipogenesis
      Gurha et al
      • Gurha P
      • Chen X
      • Lombardi R
      • Willerson JT
      • Marian AJ.
      Knockdown of plakophilin 2 downregulates miR-184 through CpG hypermethylation and suppression of the E2F1 pathway and leads to enhanced adipogenesis in vitro.
      This study validated mmu-miR-200a-3p, mmu-miR-200b-3p, and mmu-miR429-3p upregulated in cells using RT-PCR (previous results Gurha et al)HL-1 murine atrial cardiomyocytes (HL-1Pkp2-shRNA) Using low Taqman miRNAarray analysesPKP2Mechanosensing in ACM is affected through miRNA-200 family targeting in Itga1Puzzi et al
      • Puzzi L
      • Borin D
      • Gurha P
      • et al.
      Knock down of plakophillin 2 dysregulates adhesion pathway through upregulation of miR200b and alters the mechanical properties in cardiac cells.
      Two pluripotent stem cells (iPSCs) lines from an ACM differentiated to cardiomyocytes and cultured in conditioned medium from ACM patient vs healthy donorsCardiomyocytes derived from induced pluripotent cells of ACM patients Using TaqMan MicroRNA Reverse Transcription KitPKP2No differences in intracellular miRNAs. Extracellular miRNAs, miR-1 and miR-133a over-expressed and miR-302 was downregulated miR-378a was overexpressed related to adipocytesKhudiakov et al
      • Khudiakov AA
      • Smolina NA
      • Perepelina KI
      • Malashicheva AB
      • Kostareva AA.
      Extracellular microRNAs and mitochondrial DNA as potential biomarkers of arrhythmogenic cardiomyopathy.
      Arrhythmogenic phenotype in αMHC-miR130a miceTransgenic mouse model capable of inducible overexpression of miR-130a in cardiomyocytesDSC2Overexpressed miR-130a repressed DSC2 genes leading to low concentrations of this protein and desmosomal dysfunctionMazurek et al
      • Mazurek SR
      • Calway T
      • Harmon C
      • Farrell P
      • Kim GH.
      MicroRNA-130a regulation of desmocollin 2 in a novel model of arrhythmogenic cardiomyopathy.
      Nkx2.5-Cre:Pkp2shRNA and Myh6:JupTr micePKP2, JUPmiR-184Lombardi et al
      • Lombardi R
      • da Graca Cabreira-Hansen M
      • Bell A
      • Fromm RR
      • Willerson JT
      • Marian AJ.
      Nuclear plakoglobin is essential for differentiation of cardiac progenitor cells to adipocytes in arrhythmogenic right ventricular cardiomyopathy.
      A murine model with cardiomyocyte-specific overexpression of a FLAG-tagged human DSG2 was used to demonstrate 24 miRNAs dysregulatedTransgenic murine model RNA sequencingDSG2miR-217-5p and miR-708-5p were upregulated, whereas miR-499-5p was downregulated in Tg-hQ13 mice. In addition, miR-217-5p, miR-708-5p and miR-499-5p were predicted to be involved in the Wnt/β-catenin pathwayCalore et al
      • Calore M
      • Lorenzon A
      • Vitiello L
      • et al.
      A novel murine model for arrhythmogenic cardiomyopathy points to a pathogenic role of Wnt signalling and miRNA dysregulation.
      Abbreviations: ACM, arrhythmogenic myocardiopathy; DSG2, desmoglein-2; DSP, desmoplakin; HC, healthy controls; HTX, heart transplant; iPSCs, pluripotent stem cells; IVT, idiopathic ventricular tachycardia; NGS, next-generation sequencing; PKP2, plakophilin-2; RT-PCR, reverse-transcription polymerase chain reaction.

      MiRNAs in ACM cellular models

      In 2016, Gurha et al
      • Gurha P
      • Chen X
      • Lombardi R
      • Willerson JT
      • Marian AJ.
      Knockdown of plakophilin 2 downregulates miR-184 through CpG hypermethylation and suppression of the E2F1 pathway and leads to enhanced adipogenesis in vitro.
      performed the expression profile of miRNAs in 2 independent PKP2-deficient HL-1 cell (HL-1PKP2-shRNA) lines. Among the 59 miRNAs differentially expressed, miR-487b and 2 members of the miR-200 family (miR-200b and miR-429) were the most upregulated, whereas miR-184 and miR-881 were the most downregulated miRNAs. Consistent with the previous results, a study demonstrated by RT-qPCR that those 3 members of the miR-200 family, namely miR-200a-3p, miR-200b-3p, and miR429-3p, were upregulated in HL-1PKP2-shRNA cells.
      • Puzzi L
      • Borin D
      • Gurha P
      • et al.
      Knock down of plakophillin 2 dysregulates adhesion pathway through upregulation of miR200b and alters the mechanical properties in cardiac cells.
      However, only miR-200b inhibition fully restored the normal phenotype of HL-1PKP2-shRNA cells.
      • Puzzi L
      • Borin D
      • Gurha P
      • et al.
      Knock down of plakophillin 2 dysregulates adhesion pathway through upregulation of miR200b and alters the mechanical properties in cardiac cells.
      The expression of miR-184 was validated in the heart tissue of 2 ACM mouse models (Nkx2.5-Cre:Pkp2shRNA and Myh6:JupTr).
      • Lombardi R
      • da Graca Cabreira-Hansen M
      • Bell A
      • Fromm RR
      • Willerson JT
      • Marian AJ.
      Nuclear plakoglobin is essential for differentiation of cardiac progenitor cells to adipocytes in arrhythmogenic right ventricular cardiomyopathy.
      In 2019, Khudiakov et al
      • Khudiakov AA
      • Smolina NA
      • Perepelina KI
      • Malashicheva AB
      • Kostareva AA.
      Extracellular microRNAs and mitochondrial DNA as potential biomarkers of arrhythmogenic cardiomyopathy.
      reported miRNA expression analysis in a conditioned medium collected from cardiomyocytes, differentiated from pluripotent stem cells of an ACM patient carrying a pathogenic alteration in the PKP2 gene and a healthy donor. The expression of extracellular miRNAs miR-1 and miR-133a was increased in the conditioned medium obtained from ACM cells, suggesting myocardial damage. Also, higher levels of miR-21 and miR-29b-2, related to fibrosis were present in ACM supernatant, as well as miR-378a levels, a miRNA known to be expressed in adipocytes.
      • Rainer J
      • Meraviglia V
      • Blankenburg H
      • et al.
      The arrhythmogenic cardiomyopathy-specific coding and non-coding transcriptome in human cardiac stromal cells.
      Of note, analysis of intracellular miRNAs showed no difference between the expression profile of cardiomyocytes from the ACM patient and healthy donor, highlighting that the differences appear upon secretion and not inside the cells.

      MiRNAs in ACM animal models

      In concordance to Gurha et al,
      • Gurha P
      • Chen X
      • Lombardi R
      • Willerson JT
      • Marian AJ.
      Knockdown of plakophilin 2 downregulates miR-184 through CpG hypermethylation and suppression of the E2F1 pathway and leads to enhanced adipogenesis in vitro.
      the expression of miR-184 was validated in the heart tissue of 2 ACM animal models (Nkx2.5-Cre:Pkp2shRNA and Myh6:JupTr).
      • Lombardi R
      • da Graca Cabreira-Hansen M
      • Bell A
      • Fromm RR
      • Willerson JT
      • Marian AJ.
      Nuclear plakoglobin is essential for differentiation of cardiac progenitor cells to adipocytes in arrhythmogenic right ventricular cardiomyopathy.
      In a transgenic murine heart model, miR-130a was overexpressed, leading to reduced connexin-43 levels over 90% in 10 weeks of life and induced ventricular arrhythmias. Lately, connexin-43 has been confirmed to be a direct target in fibroblast and cardiomyocytes assays.
      • Osbourne A
      • Calway T
      • Broman M
      • McSharry S
      • Earley J
      • Kim GH.
      Downregulation of connexin43 by microRNA-130a in cardiomyocytes results in cardiac arrhythmias.
      Mazurek et al
      • Mazurek SR
      • Calway T
      • Harmon C
      • Farrell P
      • Kim GH.
      MicroRNA-130a regulation of desmocollin 2 in a novel model of arrhythmogenic cardiomyopathy.
      reported that transgenic mice with specific overexpression of miR-130a in adult cardiomyocytes (αMHC-miR-130a) exhibited an ACM phenotype with increased fibrosis, lipid accumulation and cardiomyocyte death. In 2019, Calore et al
      • Calore M
      • Lorenzon A
      • Vitiello L
      • et al.
      A novel murine model for arrhythmogenic cardiomyopathy points to a pathogenic role of Wnt signalling and miRNA dysregulation.
      generated a novel murine model for ACM with cardiomyocyte-specific overexpression of human WT DSG2 protein (Tg-hWT) and p.Q558X_DSG2 pathogenic variants (Tg-hQ). RNA-sequencing from myocardial samples showed the modulation of 24 miRNAs in Tg-hQ hearts compared to nontransgenic hearts, with 18 upregulated and 6 downregulated. This study identified a miRNA signature in murine ACM hearts, with miR-708-5p and miR-217-5p being the most upregulated and miR-499-5p the most downregulated miRNAs. All of them were predicted to be involved in the regulation of the Wnt/β-catenin pathway, adherence junction and gap junction, thus suggesting their potential involvement in the pathophysiology of ACM as well as their potential utility as future therapeutic targets for the disease.
      • Calore M
      • Lorenzon A
      • Vitiello L
      • et al.
      A novel murine model for arrhythmogenic cardiomyopathy points to a pathogenic role of Wnt signalling and miRNA dysregulation.

      THE POTENTIAL ROLE OF MIRNAS IN THE ACM MOLECULAR PATHWAYS

      MiRNAs are now recognized as critical regulators of diverse physiological and pathological processes; however, studies elucidating the potential biological relevance of miRNAs into the ACM pathophysiological mechanisms are still very few.

      Desmosome and GAP junctions

      Some of the desmosome and GAP junctions’ proteins have also been predicted to be directly targeted miRNAs in the context of ACM pathogenesis. Concretely, a previous study showed that luciferase reporter assays identified DSC2 as a direct target of miR-130a
      • Mazurek SR
      • Calway T
      • Harmon C
      • Farrell P
      • Kim GH.
      MicroRNA-130a regulation of desmocollin 2 in a novel model of arrhythmogenic cardiomyopathy.
      (Fig 3) and immunofluorescent staining and western blot assays showed significant reduction of DSC2 expression compared to control. On the other hand, using this mouse model, the same group previously demonstrated that miR-130a overexpression in adult cardiomyocytes promoted atrial and ventricular arrhythmias and regulated Connexin-43 expression
      • Osbourne A
      • Calway T
      • Broman M
      • McSharry S
      • Earley J
      • Kim GH.
      Downregulation of connexin43 by microRNA-130a in cardiomyocytes results in cardiac arrhythmias.
      (Fig 3).
      Fig 3
      Fig 3ACM and the 4 distinct structures in the intercalated disc involved in the pathology of this entity. Schematic flowchart of miRNAs orchestrating several mechanisms involved in ACM. Green and red means over and down expressed miRNAs. Cx43, connexion-43; DES, desmin; DSC2, Desmocollin-2; DSP, desmoplakin; DSG2, desmoglein-2; PKP2, plakophilin-2; *interaction no validated. (For interpretation of the references to color in this figure legend, the reader is referred to the Web version of this article.)

      Wnt/β-catenin pathway

      Wnt/β-catenin pathway is one of the main molecular pathways underlying the ACM cellular phenotype. Suppression of the canonical Wnt/catenin-β1 signaling has been implicated in adipogenesis, fibrogenesis, and apoptosis.
      • Austin KM
      • Trembley MA
      • Chandler SF
      • et al.
      Molecular mechanisms of arrhythmogenic cardiomyopathy.
      Also, the activation of Hippo pathway has been associated with ACM pathogenesis, due to its effect on suppressing Wnt pathway, dysregulating cell proliferation/apoptosis and enhancing adipogenesis.
      • Chen SN
      • Gurha P
      • Lombardi R
      • Ruggiero A
      • Willerson JT
      • Marian AJ.
      The hippo pathway is activated and is a causal mechanism for adipogenesis in arrhythmogenic cardiomyopathy.
      Several miRNAs have been suggested to be involved in the Wnt/β-Catenin pathway, being the miR-708-5p and miR-217-5p being the most upregulated and miR-499-5p the most downregulated miRNAs (Fig 2). All of them predicted to be involved in the regulation of Wnt/β-catenin pathway, adherence junction and gap junction.
      • Calore M
      • Lorenzon A
      • Vitiello L
      • et al.
      A novel murine model for arrhythmogenic cardiomyopathy points to a pathogenic role of Wnt signalling and miRNA dysregulation.
      On the other hand, in silico analysis showed that miR-185-5p was involved in the Wnt-Hippo pathway and cell junction process, leading to a structural functional impairment of the cardiomyocytes.
      • Sacchetto C
      • Mohseni Z
      • Colpaert RMW
      • et al.
      Circulating miR-185-5p as a potential biomarker for arrhythmogenic right ventricular cardiomyopathy.

      TGFβ signaling

      Several studies suggested that both canonical and noncanonical TGFβ signaling can have a role in inducing myocardial fibrosis in ACM. MiRNAs may play an important role in the regulation of these signaling pathways. In particular, under-expression of miR-26a and overexpression of miR-122 have been associated with angiotensin II-mediated fibrosis progression in the TGFβ-CTGF and LGR4 -β-catenin signaling pathways.
      • Song JJ
      • Yang M
      • Liu Y
      • et al.
      MicroRNA-122 aggravates angiotensin II-mediated apoptosis and autophagy imbalance in rat aortic adventitial fibroblasts via the modulation of SIRT6-elabela-ACE2 signaling.
      ,
      • Wei C
      • Kim IK
      • Kumar S
      • et al.
      NF-kappaB mediated miR-26a regulation in cardiac fibrosis.
      In these last 2 studies, Wei et al
      • Wei C
      • Kim IK
      • Kumar S
      • et al.
      NF-kappaB mediated miR-26a regulation in cardiac fibrosis.
      reported that miR-26a is regulated by Nuclear Factor κB (NF-κB) signaling, a key regulator of inflammation and oxidative stress, and interacts directly with the 3′-UTR region of collagen I and CTGF mRNAs, conferring a regulatory feedback mechanism in cardiac fibrosis. In addition, Song et al
      • Song JJ
      • Yang M
      • Liu Y
      • et al.
      MicroRNA-122 aggravates angiotensin II-mediated apoptosis and autophagy imbalance in rat aortic adventitial fibroblasts via the modulation of SIRT6-elabela-ACE2 signaling.
      also found that overexpression of miR-122, increased loss of autophagy, cell migration, apoptosis and extracellular matrix deposition, proposing it as a biomarker and therapeutic approach in cardiovascular fibrosis and related diseases. Although the role of miR-122 in the ACM pathophysiological mechanisms is still to be fully understood, a previous study in ACM cases and controls heart samples concluded that miR-122 was one of the 6 signature miRNAs (miR-122-5p, miR-133a-3p, miR-133b, miR-142-3p, miR-182-5p, and miR-183-5p) presenting high discriminatory diagnostic power in ACM.
      • Bueno Marinas M
      • Celeghin R
      • Cason M
      • et al.
      A microRNA expression profile as non-invasive biomarker in a large arrhythmogenic cardiomyopathy cohort.

      Adipogenesis

      Furthermore, miR-184 target genes associated with lipogenesis were upregulated with PKP2 knockdown, and in vitro models of aberrant miRNA-184 expression illustrated its role as a molecular switch for adipogenic gene regulation.
      • Gurha P
      • Chen X
      • Lombardi R
      • Willerson JT
      • Marian AJ.
      Knockdown of plakophilin 2 downregulates miR-184 through CpG hypermethylation and suppression of the E2F1 pathway and leads to enhanced adipogenesis in vitro.

      MAIN CHALLENGES OF MIRNA IN CLINICAL APPLICATION FOR ACM

      MiRNAs are the most widely studied among all ncRNAs due to the fact that they are easily detectable in corporal fluids and tissues, cell-type specific, and they show distinct patterns and fluctuation based on physiological or pathological stress. Therefore, miRNAs can be studied using high-throughput miRNA sequencing or by arrays. The latter or real-time PCR are a fast, low-cost and reliable technique, very useful to screen miRNAs from various corporal fluids. On the other hand, high throughput miRNA sequencing provides a more complex and unbiased analysis, including global profiles of nonannotated ncRNAs
      • Lu D
      • Thum T.
      RNA-based diagnostic and therapeutic strategies for cardiovascular disease.
      This analysis enriches the differential gene expression, alternative splicing, pathway findings and co-expression network.
      Over the past decade, miRNAs have had massively increased attention as diagnostic biomarkers in cardiovascular diseases due to evidence of their potential role in pathogenesis. Multiple studies suggest that the circulating miRNA signature reflects the physiological or pathological status of a patient, stratifying their prognosis and tailoring personalized therapies. Besides, the analysis of miRNA patterns may help to understand the molecular mechanisms underlying the progression of the disease. Indeed, as we have discussed in this review, overexpression or suppression of miRNAs can contribute to the development of distinct cardiovascular disorders, including ACM. In this sense, miRNA mimics and anti-miRNA technology have triggered enthusiasm for miRNAs as potential drug targets. However, there are barriers to the clinical application of miRNAs. These include, for instance, the instability of miRNAs and the reproducibility of the results.
      A handicap in daily practice is the absence of standardized methods, since the sample collection and storage, the total RIN value needed for an RNA analysis.
      • Robinson EL
      • Baker AH
      • Brittan M
      • et al.
      Dissecting the transcriptome in cardiovascular disease.
      There are no accurate, consistent and robust methods for detecting and measuring circulating miRNA, highlighting the importance of standardizing guidelines as well as sample preparation and quantification methods.
      • Felekkis K
      • Papaneophytou C.
      Challenges in using circulating micro-RNAs as biomarkers for cardiovascular diseases.
      In this regard, it has been shown that different methods of sample collection and storage result in a different quality of isolated RNA. Thus, the impact of RNA degradation and DNA contamination and, more importantly, low-input RNA analysis may influence the final results. Moreover, total RNA is frequently prepared from samples that were archived for various periods in frozen tissue banks. Unfortunately, the quality of samples stored in tissue banks is variable and RNA integrity is not always preserved, which will impact the reliability of the results of the analysis.
      • Ibberson D
      • Benes V
      • Muckenthaler MU
      • Castoldi M.
      RNA degradation compromises the reliability of microRNA expression profiling.
      Besides, the differences between RNA isolation protocols affect the relative concentrations of miRNAs and limit the experimental reproducibility. On the other hand, the use of miRNAs to untangle their role in the pathophysiology as key regulators is limited by the experimental design, using animal or cellular models and their biological differences, and the inhomogeneity of miRNA depending on their origin. Human samples for any cardiovascular disease and ACM are usually restricted. However, even when they are accessible, several factors may influence the transcriptomic results, such as post-mortem interval of time, temperature, and handling protocols.
      The lack of consensus regarding methodologies used for miRNA quantification is one of the main limiting factors. Three principal methods are used to measure the expression levels of miRNAs: real-time reverse transcription-PCR (qRT-PCR),
      • Chen C
      • Ridzon DA
      • Broomer AJ
      • et al.
      Real-time quantification of microRNAs by stem-loop RT-PCR.
      ,
      • Shi R
      • Chiang VL.
      Facile means for quantifying microRNA expression by real-time PCR.
      microarray hybridization,
      • Yin BC
      • Li H
      • Ye BC.
      A dual-probe hybridization method for reducing variability in single nucleotide polymorphism analysis with oligonucleotide microarrays.
      ,
      • Li W
      • Ruan K.
      MicroRNA detection by microarray.
      and next-generation sequencing (NGS),
      • Hafner M
      • Landgraf P
      • Ludwig J
      • et al.
      Identification of microRNAs and other small regulatory RNAs using cDNA library sequencing.
      and each method has its limitations, thus affecting to the reproducibility of the results. Regarding circulating miRNAs, the lack of a universally invariant reference miRNA for normalization has become a serious challenge. Circulating miRNAs do not confirm their heart origin but show more specificity and sensitivity than other biomarkers. Nevertheless, consistent multicentric studies are required to achieve the size of an ACM sample that allows the establishment of a panel of miRNAs as biomarkers for ACM.
      • Benesova S
      • Kubista M
      • Valihrach L.
      Small RNA-sequencing: approaches and considerations for miRNA analysis.
      Intrinsic features linked to this entity, such as low prevalence, different penetrance, variety of genotypes and phenotypes, as well as segregation make it difficult to achieve this goal.
      Finally, it is widely accepted the progressively increased use of miRNA as biomarkers in clinical practice. In this sense, the miRNA signature described by Bueno et al,
      • Bueno Marinas M
      • Celeghin R
      • Cason M
      • et al.
      A microRNA expression profile as non-invasive biomarker in a large arrhythmogenic cardiomyopathy cohort.
      and the miR-185-5p reported by Sacchetto et al
      • Sacchetto C
      • Mohseni Z
      • Colpaert RMW
      • et al.
      Circulating miR-185-5p as a potential biomarker for arrhythmogenic right ventricular cardiomyopathy.
      have been proposed as promising miRNA for early diagnosis. However, all published studies using human samples have identified miRNA in patients showing ACM phenotypes but not necessarily with an early stage of the disease as well as, even before disease. Thus, the current knowledge in miRNA is probably not yet enough to propose miRNA biomarkers for early diagnosis of ACM.

      CONCLUSION

      In recent years, several data highlight the role of microRNAs in cellular processes of human physiology, despite the fact that a large part of mechanisms still remain to be clarified. Their potential role as biomarkers in different diseases has also been suggested in studies performed in different models, but no definite agreement among this wide variety of experimental studies exists to date, including in ACM. Variable expressivity and incomplete penetrance, hallmarks of this entity, even in families carrying a deleterious variant responsible for the disease, highlight the potential role of miRNAs as biomarkers.

      ACKNOWLEDGMENTS

      Conflicts of interest: All authors have reported that they have no relationships relevant to the contents of this paper to disclose.
      This work was supported by Obra Social “La Caixa Foundation” (LCF/PR/GN19/50320002), Sociedad Española Cardiología (SEC) “Proyecto Investigación Básica Cardiología 2020 (SEC/FEC-INV-BAS 20/003),” Instituto de Salud Carlos III (FIS PI22/00422) and Foundation Progreso y Salud PEER (2020-019), grants in the framework of the European Regional Development Fund (ERDF) Integrated Territorial Initiative (ITI PI0017-2019). CIBERCV is an initiative of the ISCIII, Spanish Ministry of Economy and Competitiveness. Funders had no role in study design, data collection, data analysis, interpretation, or writing of the report.
      Author contributions are as follows: M. Alcalde, F. Bonet, J. Córdoba-Caballero, G. Sarquella-Brugada: data curation, formal analysis, investigation, methodology, validation, writing—original draft. J. A. Ranea, M. Vallverdú-Prats, R. Brugada, V. Meraviglia, M. Bellin: data curation, formal analysis, investigation, validation. R. Toro, O. Campuzano: conceptualization, data curation, formal analysis, investigation, methodology, project administration, supervision, validation, writing—review & editing. All authors certify that they have participated sufficiently in this work to take public responsibility for the content. All authors have read the journal's authorship agreement, reviewed, and approved the manuscript.

      REFERENCES

        • Kohela A
        • van Rooij E.
        Fibro-fatty remodelling in arrhythmogenic cardiomyopathy.
        Basic Res Cardiol. 2022; 117: 22
        • Wilde AAM
        • Semsarian C
        • Marquez MF
        • et al.
        European Heart Rhythm Association (EHRA)/Heart Rhythm Society (HRS)/Asia Pacific Heart Rhythm Society (APHRS)/Latin American Heart Rhythm Society (LAHRS) expert consensus statement on the state of genetic testing for cardiac diseases.
        Heart Rhythm. 2022; 19: e1-e60
        • Grassi S
        • Campuzano O
        • Coll M
        • et al.
        Update on the diagnostic pitfalls of autopsy and post-mortem genetic testing in cardiomyopathies.
        Int J Mol Sci. 2021; 22: 4124
        • D'Ascenzi F
        • Valentini F
        • Pistoresi S
        • et al.
        Causes of sudden cardiac death in young athletes and non-athletes: systematic review and meta-analysis: sudden cardiac death in the young.
        Trends Cardiovasc Med. 2022; 32: 299-308
        • Krahn AD
        • Wilde AAM
        • Calkins H
        • et al.
        Arrhythmogenic right ventricular cardiomyopathy.
        JACC Clin Electrophysiol. 2022; 8: 533-553
        • Gerull B
        • Brodehl A.
        Insights into genetics and pathophysiology of arrhythmogenic cardiomyopathy.
        Curr Heart Fail Rep. 2021; 18: 378-390
        • Cohen MI
        • Atkins MB.
        Arrhythmogenic right ventricular cardiomyopathy in the pediatric population.
        Curr Opin Cardiol. 2022; 37: 99-108
        • Odak M
        • Douedi S
        • Mararenko A
        • et al.
        Arrhythmogenic right ventricular cardiomyopathy: the role of genetics in diagnosis, management, and screening.
        Cardiol Res. 2022; 13: 177-184
        • Graziano F
        • Zorzi A
        • Cipriani A
        • et al.
        The 2020 “Padua Criteria” for diagnosis and phenotype characterization of arrhythmogenic cardiomyopathy in clinical practice.
        J Clin Med. 2022; 11: 279
        • Reichart D
        • Lindberg EL
        • Maatz H
        • et al.
        Pathogenic variants damage cell composition and single cell transcription in cardiomyopathies.
        Science. 2022; 377: eabo1984
        • de Brouwer R
        • Bosman LP
        • Gripenstedt S
        • et al.
        Value of genetic testing in the diagnosis and risk stratification of arrhythmogenic right ventricular cardiomyopathy.
        Heart Rhythm. 2022; 19: 1659-1665
        • Carvajal-Huerta L.
        Epidermolytic palmoplantar keratoderma with woolly hair and dilated cardiomyopathy.
        J Am Acad Dermatol. 1998; 39: 418-421
        • Protonotarios N
        • Tsatsopoulou A.
        Naxos disease and Carvajal syndrome: cardiocutaneous disorders that highlight the pathogenesis and broaden the spectrum of arrhythmogenic right ventricular cardiomyopathy.
        Cardiovasc Pathol. 2004; 13: 185-194
        • Simpson MA
        • Mansour S
        • Ahnood D
        • et al.
        Homozygous mutation of desmocollin-2 in arrhythmogenic right ventricular cardiomyopathy with mild palmoplantar keratoderma and woolly hair.
        Cardiology. 2009; 113: 28-34
        • Bourfiss M
        • van Vugt M
        • Alasiri AI
        • et al.
        Prevalence and disease expression of pathogenic and likely pathogenic variants associated with inherited cardiomyopathies in the general population.
        Circ Genom Precis Med. 2022; 15e003704
        • Vallverdu-Prats M
        • Alcalde M
        • Sarquella-Brugada G
        • et al.
        Rare variants associated with arrhythmogenic cardiomyopathy: reclassification five years later.
        J Pers Med. 2021; 11: 162
        • Austin KM
        • Trembley MA
        • Chandler SF
        • et al.
        Molecular mechanisms of arrhythmogenic cardiomyopathy.
        Nat Rev Cardiol. 2019; 16: 519-537
        • Kirchhof P
        • Fabritz L
        • Zwiener M
        • et al.
        Age- and training-dependent development of arrhythmogenic right ventricular cardiomyopathy in heterozygous plakoglobin-deficient mice.
        Circulation. 2006; 114: 1799-1806
        • Hariharan V
        • Asimaki A
        • Michaelson JE
        • et al.
        Arrhythmogenic right ventricular cardiomyopathy mutations alter shear response without changes in cell-cell adhesion.
        Cardiovasc Res. 2014; 104: 280-289
        • Garcia-Gras E
        • Lombardi R
        • Giocondo MJ
        • et al.
        Suppression of canonical Wnt/beta-catenin signaling by nuclear plakoglobin recapitulates phenotype of arrhythmogenic right ventricular cardiomyopathy.
        J Clin Invest. 2006; 116: 2012-2021
        • Asimaki A
        • Kapoor S
        • Plovie E
        • et al.
        Identification of a new modulator of the intercalated disc in a zebrafish model of arrhythmogenic cardiomyopathy.
        Sci Transl Med. 2014; 6: 240ra74
        • Rouhi L
        • Fan S
        • Cheedipudi SM
        • et al.
        The EP300/TP53 pathway, a suppressor of the Hippo and canonical WNT pathways, is activated in human hearts with arrhythmogenic cardiomyopathy in the absence of overt heart failure.
        Cardiovasc Res. 2022; 118: 1466-1478
        • Banerjee I
        • Yekkala K
        • Borg TK
        • Baudino TA.
        Dynamic interactions between myocytes, fibroblasts, and extracellular matrix.
        Ann N Y Acad Sci. 2006; 1080: 76-84
        • Dobaczewski M
        • Gonzalez-Quesada C
        • Frangogiannis NG.
        The extracellular matrix as a modulator of the inflammatory and reparative response following myocardial infarction.
        J Mol Cell Cardiol. 2010; 48: 504-511
        • Li CL
        • Leng JH
        • Li MH
        • Shi JH
        • Jia SZ
        • Lang JH.
        Expressions and roles of TGFbeta/Smad signal pathway in peritoneum of endometriosis.
        Zhonghua Fu Chan Ke Za Zhi. 2011; 46: 826-830
        • Maione AS
        • Pilato CA
        • Casella M
        • et al.
        Fibrosis in arrhythmogenic cardiomyopathy: the phantom thread in the fibro-adipose tissue.
        Front Physiol. 2020; 11: 279
        • Meier H
        • Bullinger J
        • Deten A
        • et al.
        Tissue inhibitor of matrix metalloproteinase-1 in norepinephrine-induced remodeling of the mouse heart.
        Cell Physiol Biochem. 2007; 20: 825-836
        • Jain M
        • Rivera S
        • Monclus EA
        • et al.
        Mitochondrial reactive oxygen species regulate transforming growth factor-beta signaling.
        J Biol Chem. 2013; 288: 770-777
        • Dubash AD
        • Kam CY
        • Aguado BA
        • et al.
        Plakophilin-2 loss promotes TGF-beta1/p38 MAPK-dependent fibrotic gene expression in cardiomyocytes.
        J Cell Biol. 2016; 212: 425-438
        • Thiene G
        • Corrado D
        • Nava A
        • et al.
        Right ventricular cardiomyopathy: is there evidence of an inflammatory aetiology?.
        Eur Heart J. 1991; 12: 22-25
        • Basso C
        • Thiene G
        • Corrado D
        • Angelini A
        • Nava A
        • Valente M.
        Arrhythmogenic right ventricular cardiomyopathy. Dysplasia, dystrophy, or myocarditis?.
        Circulation. 1996; 94: 983-991
        • Corrado D
        • Basso C
        • Thiene G
        • et al.
        Spectrum of clinicopathologic manifestations of arrhythmogenic right ventricular cardiomyopathy/dysplasia: a multicenter study.
        J Am Coll Cardiol. 1997; 30: 1512-1520
        • Calabrese F
        • Basso C
        • Carturan E
        • Valente M
        • Thiene G.
        Arrhythmogenic right ventricular cardiomyopathy/dysplasia: is there a role for viruses?.
        Cardiovasc Pathol. 2006; 15: 11-17
        • Campuzano O
        • Alcalde M
        • Iglesias A
        • et al.
        Arrhythmogenic right ventricular cardiomyopathy: severe structural alterations are associated with inflammation.
        J Clin Pathol. 2012; 65: 1077-1083
        • Lota AS
        • Hazebroek MR
        • Theotokis P
        • et al.
        Genetic architecture of acute myocarditis and the overlap with inherited cardiomyopathy.
        Circulation. 2022; 146: 1123-1134
        • Seidel F
        • Laser KT
        • Klingel K
        • et al.
        Pathogenic variants in cardiomyopathy disorder genes underlie pediatric myocarditis-further impact of heterozygous immune disorder gene variants?.
        J Cardiovasc Dev Dis. 2022; 9: 216
        • Kissopoulou A
        • Fernlund E
        • Holmgren C
        • et al.
        Monozygotic twins with myocarditis and a novel likely pathogenic desmoplakin gene variant.
        ESC Heart Fail. 2020; 7: 1210-1216
        • Tiron C
        • Campuzano O
        • Fernandez-Falgueras A
        • et al.
        Prevalence of pathogenic variants in cardiomyopathy-associated genes in myocarditis.
        Circ Genom Precis Med. 2022; 15e003408
        • Campuzano O
        • Fernandez-Falgueras A
        • Sarquella-Brugada G
        • et al.
        A genetically vulnerable myocardium may predispose to myocarditis.
        J Am Coll Cardiol. 2015; 66: 2913-2914
        • Meraviglia V
        • Alcalde M
        • Campuzano O
        • Bellin M.
        Inflammation in the pathogenesis of arrhythmogenic cardiomyopathy: secondary event or active driver?.
        Front Cardiovasc Med. 2021; 8784715
        • Lombardi R
        • Dong J
        • Rodriguez G
        • et al.
        Genetic fate mapping identifies second heart field progenitor cells as a source of adipocytes in arrhythmogenic right ventricular cardiomyopathy.
        Circ Res. 2009; 104: 1076-1084
        • Moses KA
        • DeMayo F
        • Braun RM
        • Reecy JL
        • Schwartz RJ.
        Embryonic expression of an Nkx2-5/Cre gene using ROSA26 reporter mice.
        Genesis. 2001; 31: 176-180
        • Verzi MP
        • McCulley DJ
        • De Val S
        • Dodou E
        • Black BL.
        The right ventricle, outflow tract, and ventricular septum comprise a restricted expression domain within the secondary/anterior heart field.
        Dev Biol. 2005; 287: 134-145
        • Zhou B
        • von Gise A
        • Ma Q
        • Rivera-Feliciano J
        • Pu WT.
        Nkx2-5- and Isl1-expressing cardiac progenitors contribute to proepicardium.
        Biochem Biophys Res Commun. 2008; 375: 450-453
        • Dorn T
        • Kornherr J
        • Parrotta EI
        • et al.
        Interplay of cell-cell contacts and RhoA/MRTF-A signaling regulates cardiomyocyte identity.
        EMBO J. 2018; 37: e98133
        • Laudes M.
        Role of WNT signalling in the determination of human mesenchymal stem cells into preadipocytes.
        J Mol Endocrinol. 2011; 46: R65-R72
        • Gao S
        • Puthenvedu D
        • Lombardi R
        • Chen SN.
        Established and emerging mechanisms in the pathogenesis of arrhythmogenic cardiomyopathy: a multifaceted disease.
        Int J Mol Sci. 2020; 21
        • Willert K
        • Shibamoto S
        • Nusse R.
        Wnt-induced dephosphorylation of axin releases beta-catenin from the axin complex.
        Genes Dev. 1999; 13: 1768-1773
        • Chen S
        • Guttridge DC
        • You Z
        • et al.
        Wnt-1 signaling inhibits apoptosis by activating beta-catenin/T cell factor-mediated transcription.
        J Cell Biol. 2001; 152: 87-96
        • Hoorntje ET
        • Te Rijdt WP
        • James CA
        • et al.
        Arrhythmogenic cardiomyopathy: pathology, genetics, and concepts in pathogenesis.
        Cardiovasc Res. 2017; 113: 1521-1531
        • Niehrs C.
        The complex world of WNT receptor signalling.
        Nat Rev Mol Cell Biol. 2012; 13: 767-779
        • Kostis WJ
        • Tedford RJ
        • Miller DL
        • Schulman SP
        • Tomaselli GF.
        Troponin-I elevation in a young man with arrhythmogenic right ventricular dysplasia/cardiomyopathy.
        J Interv Card Electrophysiol. 2008; 22: 49-53
        • Wei YJ
        • Huang YX
        • Shen Y
        • et al.
        Proteomic analysis reveals significant elevation of heat shock protein 70 in patients with chronic heart failure due to arrhythmogenic right ventricular cardiomyopathy.
        Mol Cell Biochem. 2009; 332: 103-111
        • Hong TT
        • Cogswell R
        • James CA
        • et al.
        Plasma BIN1 correlates with heart failure and predicts arrhythmia in patients with arrhythmogenic right ventricular cardiomyopathy.
        Heart Rhythm. 2012; 9: 961-967
        • Matsuo K
        • Nishikimi T
        • Yutani C
        • et al.
        Diagnostic value of plasma levels of brain natriuretic peptide in arrhythmogenic right ventricular dysplasia.
        Circulation. 1998; 98: 2433-2440
        • Bueno Marinas M
        • Celeghin R
        • Cason M
        • et al.
        A microRNA expression profile as non-invasive biomarker in a large arrhythmogenic cardiomyopathy cohort.
        Int J Mol Sci. 2020; 21: 1536
        • Cheng H
        • Lu M
        • Hou C
        • et al.
        Relation between N-terminal pro-brain natriuretic peptide and cardiac remodeling and function assessed by cardiovascular magnetic resonance imaging in patients with arrhythmogenic right ventricular cardiomyopathy.
        Am J Cardiol. 2015; 115: 341-347
        • Akdis D
        • Saguner AM
        • Burri H
        • et al.
        Clinical predictors of left ventricular involvement in arrhythmogenic right ventricular cardiomyopathy.
        Am Heart J. 2020; 223: 34-43
        • Stadiotti I
        • Pompilio G
        • Maione AS
        • Pilato CA
        • D'Alessandra Y
        • Sommariva E
        Arrhythmogenic cardiomyopathy: what blood can reveal?.
        Heart Rhythm. 2019; 16: 470-477
        • Holte E
        • Kleveland O
        • Ueland T
        • et al.
        Effect of interleukin-6 inhibition on coronary microvascular and endothelial function in myocardial infarction.
        Heart. 2017; 103: 1521-1527
        • Oz F
        • Onur I
        • Elitok A
        • et al.
        Galectin-3 correlates with arrhythmogenic right ventricular cardiomyopathy and predicts the risk of ventricular-arrhythmias in patients with implantable defibrillators.
        Acta Cardiol. 2017; 72: 453-459
        • Francia P
        • Adduci C
        • Semprini L
        • et al.
        Osteopontin and galectin-3 predict the risk of ventricular tachycardia and fibrillation in heart failure patients with implantable defibrillators.
        J Cardiovasc Electrophysiol. 2014; 25: 609-616
        • Campian ME
        • Verberne HJ
        • Hardziyenka M
        • et al.
        Assessment of inflammation in patients with arrhythmogenic right ventricular cardiomyopathy/dysplasia.
        Eur J Nucl Med Mol Imaging. 2010; 37: 2079-2085
        • Asimaki A
        • Tandri H
        • Duffy ER
        • et al.
        Altered desmosomal proteins in granulomatous myocarditis and potential pathogenic links to arrhythmogenic right ventricular cardiomyopathy.
        Circ Arrhythm Electrophysiol. 2011; 4: 743-752
        • Bonny A
        • Lellouche N
        • Ditah I
        • et al.
        C-reactive protein in arrhythmogenic right ventricular dysplasia/cardiomyopathy and relationship with ventricular tachycardia.
        Cardiol Res Pract. 2010; 2010: 919783
        • Marcus FI
        • McKenna WJ
        • Sherrill D
        • et al.
        Diagnosis of arrhythmogenic right ventricular cardiomyopathy/dysplasia: proposed modification of the task force criteria.
        Circulation. 2010; 121: 1533-1541
        • Gabriel AF
        • Costa MC
        • Enguita FJ.
        Interactions among regulatory non-coding RNAs involved in cardiovascular diseases.
        Adv Exp Med Biol. 2020; 1229: 79-104
        • Jiang W
        • Agrawal DK
        • Boosani CS.
        Non-coding RNAs as epigenetic gene regulators in cardiovascular diseases.
        Adv Exp Med Biol. 2020; 1229: 133-148
        • van Rooij E
        • Sutherland LB
        • Liu N
        • et al.
        A signature pattern of stress-responsive microRNAs that can evoke cardiac hypertrophy and heart failure.
        Proc Natl Acad Sci U S A. 2006; 103: 18255-18260
        • Calderon-Dominguez M
        • Mangas A
        • Belmonte T
        • Quezada-Feijoo M
        • Ramos M
        • Toro R.
        Ischemic dilated cardiomyopathy pathophysiology through microRNA-16-5p.
        Rev Esp Cardiol (Engl Ed). 2021; 74: 740-749
        • Small EM
        • Olson EN.
        Pervasive roles of microRNAs in cardiovascular biology.
        Nature. 2011; 469: 336-342
        • Beermann J
        • Piccoli MT
        • Viereck J
        • Thum T.
        Non-coding RNAs in development and disease: background, mechanisms, and therapeutic approaches.
        Physiol Rev. 2016; 96: 1297-1325
        • Liu N
        • Olson EN.
        MicroRNA regulatory networks in cardiovascular development.
        Dev Cell. 2010; 18: 510-525
        • Gu W
        • Xu Y
        • Xie X
        • Wang T
        • Ko JH
        • Zhou T.
        The role of RNA structure at 5′ untranslated region in microRNA-mediated gene regulation.
        RNA. 2014; 20: 1369-1375
        • Brummer A
        • Hausser J.
        MicroRNA binding sites in the coding region of mRNAs: extending the repertoire of post-transcriptional gene regulation.
        Bioessays. 2014; 36: 617-626
        • Bartel DP.
        MicroRNAs: target recognition and regulatory functions.
        Cell. 2009; 136: 215-233
        • Hayder H
        • O'Brien J
        • Nadeem U
        • Peng C.
        MicroRNAs: crucial regulators of placental development.
        Reproduction. 2018; 155: R259-R271
        • Okamura K
        • Hagen JW
        • Duan H
        • Tyler DM
        • Lai EC.
        The mirtron pathway generates microRNA-class regulatory RNAs in Drosophila.
        Cell. 2007; 130: 89-100
        • Freedman JE
        • Gerstein M
        • Mick E
        • et al.
        Diverse human extracellular RNAs are widely detected in human plasma.
        Nat Commun. 2016; 7: 11106
        • Sommariva E
        • D'Alessandra Y
        • Farina FM
        • et al.
        MiR-320a as a potential novel circulating biomarker of arrhythmogenic cardiomyopathy.
        Sci Rep. 2017; 7: 4802
        • Hamam D
        • Ali D
        • Vishnubalaji R
        • et al.
        microRNA-320/RUNX2 axis regulates adipocytic differentiation of human mesenchymal (skeletal) stem cells.
        Cell Death Dis. 2014; 5: e1499
        • Yamada S
        • Hsiao YW
        • Chang SL
        • et al.
        Circulating microRNAs in arrhythmogenic right ventricular cardiomyopathy with ventricular arrhythmia.
        Europace. 2018; 20: f37-f45
        • Thum T
        • Galuppo P
        • Wolf C
        • et al.
        MicroRNAs in the human heart: a clue to fetal gene reprogramming in heart failure.
        Circulation. 2007; 116: 258-267
        • Zhang H
        • Liu S
        • Dong T
        • et al.
        Profiling of differentially expressed microRNAs in arrhythmogenic right ventricular cardiomyopathy.
        Sci Rep. 2016; 6: 28101
        • Gurha P
        • Chen X
        • Lombardi R
        • Willerson JT
        • Marian AJ.
        Knockdown of plakophilin 2 downregulates miR-184 through CpG hypermethylation and suppression of the E2F1 pathway and leads to enhanced adipogenesis in vitro.
        Circ Res. 2016; 119: 731-750
        • Puzzi L
        • Borin D
        • Gurha P
        • et al.
        Knock down of plakophillin 2 dysregulates adhesion pathway through upregulation of miR200b and alters the mechanical properties in cardiac cells.
        Cells. 2019; 8: 1639
        • Lombardi R
        • da Graca Cabreira-Hansen M
        • Bell A
        • Fromm RR
        • Willerson JT
        • Marian AJ.
        Nuclear plakoglobin is essential for differentiation of cardiac progenitor cells to adipocytes in arrhythmogenic right ventricular cardiomyopathy.
        Circ Res. 2011; 109: 1342-1353
        • Khudiakov AA
        • Smolina NA
        • Perepelina KI
        • Malashicheva AB
        • Kostareva AA.
        Extracellular microRNAs and mitochondrial DNA as potential biomarkers of arrhythmogenic cardiomyopathy.
        Biochemistry (Mosc). 2019; 84: 272-282
        • Rainer J
        • Meraviglia V
        • Blankenburg H
        • et al.
        The arrhythmogenic cardiomyopathy-specific coding and non-coding transcriptome in human cardiac stromal cells.
        BMC Genom. 2018; 19: 491
        • Osbourne A
        • Calway T
        • Broman M
        • McSharry S
        • Earley J
        • Kim GH.
        Downregulation of connexin43 by microRNA-130a in cardiomyocytes results in cardiac arrhythmias.
        J Mol Cell Cardiol. 2014; 74: 53-63
        • Calore M
        • Lorenzon A
        • Vitiello L
        • et al.
        A novel murine model for arrhythmogenic cardiomyopathy points to a pathogenic role of Wnt signalling and miRNA dysregulation.
        Cardiovasc Res. 2019; 115: 739-751
        • Mazurek SR
        • Calway T
        • Harmon C
        • Farrell P
        • Kim GH.
        MicroRNA-130a regulation of desmocollin 2 in a novel model of arrhythmogenic cardiomyopathy.
        Microrna. 2017; 6: 143-150
        • Chen SN
        • Gurha P
        • Lombardi R
        • Ruggiero A
        • Willerson JT
        • Marian AJ.
        The hippo pathway is activated and is a causal mechanism for adipogenesis in arrhythmogenic cardiomyopathy.
        Circ Res. 2014; 114: 454-468
        • Sacchetto C
        • Mohseni Z
        • Colpaert RMW
        • et al.
        Circulating miR-185-5p as a potential biomarker for arrhythmogenic right ventricular cardiomyopathy.
        Cells. 2021; 10: 2578
        • Song JJ
        • Yang M
        • Liu Y
        • et al.
        MicroRNA-122 aggravates angiotensin II-mediated apoptosis and autophagy imbalance in rat aortic adventitial fibroblasts via the modulation of SIRT6-elabela-ACE2 signaling.
        Eur J Pharmacol. 2020; 883173374
        • Wei C
        • Kim IK
        • Kumar S
        • et al.
        NF-kappaB mediated miR-26a regulation in cardiac fibrosis.
        J Cell Physiol. 2013; 228: 1433-1442
        • Lu D
        • Thum T.
        RNA-based diagnostic and therapeutic strategies for cardiovascular disease.
        Nat Rev Cardiol. 2019; 16: 661-674
        • Robinson EL
        • Baker AH
        • Brittan M
        • et al.
        Dissecting the transcriptome in cardiovascular disease.
        Cardiovasc Res. 2022; 118: 1004-1019
        • Felekkis K
        • Papaneophytou C.
        Challenges in using circulating micro-RNAs as biomarkers for cardiovascular diseases.
        Int J Mol Sci. 2020; 21: 561
        • Ibberson D
        • Benes V
        • Muckenthaler MU
        • Castoldi M.
        RNA degradation compromises the reliability of microRNA expression profiling.
        BMC Biotechnol. 2009; 9: 102
        • Chen C
        • Ridzon DA
        • Broomer AJ
        • et al.
        Real-time quantification of microRNAs by stem-loop RT-PCR.
        Nucl Acids Res. 2005; 33: e179
        • Shi R
        • Chiang VL.
        Facile means for quantifying microRNA expression by real-time PCR.
        Biotechniques. 2005; 39: 519-525
        • Yin BC
        • Li H
        • Ye BC.
        A dual-probe hybridization method for reducing variability in single nucleotide polymorphism analysis with oligonucleotide microarrays.
        Anal Biochem. 2008; 383: 270-278
        • Li W
        • Ruan K.
        MicroRNA detection by microarray.
        Anal Bioanal Chem. 2009; 394: 1117-1124
        • Hafner M
        • Landgraf P
        • Ludwig J
        • et al.
        Identification of microRNAs and other small regulatory RNAs using cDNA library sequencing.
        Methods. 2008; 44: 3-12
        • Benesova S
        • Kubista M
        • Valihrach L.
        Small RNA-sequencing: approaches and considerations for miRNA analysis.
        Diagnostics (Basel). 2021; 11: 964