Open access peer-reviewed chapter

Emerging Hallmarks of Mitochondrial Biochemistry in Cardiac Trabecular Morphogenesis and Left Ventricular Noncompaction (LVNC)

Written By

Gowthami Mahendran and Margaret A. Schwarz

Submitted: 24 June 2022 Reviewed: 22 November 2022 Published: 20 December 2022

DOI: 10.5772/intechopen.109098

From the Edited Volume

New Insights on Cardiomyopathy

Edited by Sameh M. Said

Chapter metrics overview

86 Chapter Downloads

View Full Metrics

Abstract

Functioning as a pivotal platform for energy production and transduction, mitochondria generate ATP to meet the dynamic demands of embryonic development. Consequently, disruption or alteration in mitochondrial activity influences not only cellular status, but also can impact organ formation. Disrupted mitochondrial performance not only impairs cardiovascular function but can also disrupt cardiac maturation through prevention of the myocardium’s transition between the trabeculation to the compaction phase. During embryonic development, proliferating cardiomyocytes create a trabecular mesh network. Gradual compaction of this network transforms the intra-trabecular spaces into the capillaries of the coronary circulation. Achievement of functional compaction and ultimately normal cardiac function is dependent in part on mitochondrial well-being with failure to complete remodeling of the inner trabecular layer contributing to disrupted endocardial vasculature and fibrosis, left ventricular noncompaction (LVNC). LVNC, commonly associated with mitochondrial genetic alterations, is speculated to occur due to an interruption during the process of compaction at the early developmental stages of the left ventricle (LV). Mitochondrial mutations, remain the common etiology of LVNC with a wide spectrum of these genes associated with other cardiomyopathies related to LVNC. Understanding the impact that mitochondrial genetic alterations have on the evolution of cardiac noncompaction could provide new treatment opportunities.

Keywords

  • trabeculation
  • mitochondria
  • LVNC
  • mutations
  • cardiomyopathy

1. Introduction

Cardiac development consists of four stages including embryonic cardiac chamber maturation [Carnegie stage (CS) 11 in humans and embryonic day (E) 8.5 in mice], trabeculation (at CS 12 in humans and E9.5 in mice), compaction (CS 22 in humans and E14.5 in mice) and cardiomyocyte proliferation (E11-E17.5 in mice). Originating from embryonic mesodermal germ layer cells that differentiate into mesothelium, endothelium, and myocardium followed by gastrulation, the exterior lining of the heart is made up of mesothelial pericardium. Whereas the interior lining of the heart, lymphatic, and blood vessels, arise from endothelium. During cardiac development, the atrial septation (primary atrial septum formation) begins at E10.5 to E13.5 in mice, which is comparable to Carnegie stages (CS 14 to 18) in humans or estimated gestational age (EGA) of 6 6/7–8 weeks. The ventricular septation proceeds from E11.5 to E13.5 in mice and EGA 8–9 1/7 weeks (CS 18–22) in human fetuses. Outflow septation, the transformation of a right ventricle (RV) single outflow tract into pulmonary and aortic arteries originating from the right and left ventricles, starts at E11.5–E13.5 in the mouse and EGA 7 3/7–8 weeks (CS 16–18) in humans [1]. While the separation of atrioventricular (AV) valves from endocardial cushions at the center of cardiac loop occurs at 9 1/7 weeks (CS 22). By the end of 9 1/7 weeks (CS22), all of the major structures of the heart are formed, with the average spanning period of EGA 6 4/7–9 3/7 weeks [2, 3, 4].

During cardiac development, trabeculation followed by a compaction step (CS 12 to CS 22), is vital in the developmental stages of humans (Figure 1). Emerging from the developing ventricular wall and stretching into the ventricular lumen, trabeculae projections consist of cardiomyocytes lined by an endothelial layer of the endocardium and are an important element of ordered ventricular formation. Any defects during trabeculation, compaction of remodeling junctions, or cardiac chamber maturation, could lead to the inhibition of well compacted myocardium with persisting trabeculations resulting in the condition left ventricular noncompaction (LVNC).

Figure 1.

Progression of left ventricular trabeculation to compaction: Trabeculation and compaction of endocardial cells (yellow), cardiomyocytes (orange in left image, red in right image), cardiac jelly (purple), epicardial cells (blue) of a LV followed by the maturation step in mouse and human are shown (figure adapted and modified from Zhang et al. [5]).

The trabeculation process is further defined by three sequential steps resulting in fusion with myocardium layer that is compacted and progression into a mature thickened ventricular wall. Initially, the trabecular ridges start to emerge while the myocardial projections progress into the lumen. This is followed by the expansion of trabeculae projections, creating a trabeculae network. In the final remodeling step of trabeculae formation, trabeculae growth ceases and they compact together contributing to ventricular radial thickening [6, 7]. Importantly, the LV of a healthy heart has three distinct trabeculation types (hypertrophic, fibrotic or both) [8] and is less trabeculated than RV [9]. Upon stimulation of ventricular trabeculations during the compaction phase of embryonic development, the growth of trabecula ceases and the ends of the trabecula thicken, with the spaces in between the trabecular buds developing into capillaries [10, 11]. The persistence of a trabecular mesh caused by myocardial noncompaction can occur due to irregularities arising in the maturation process of the cardiac chamber. However, the step of remodeling ventricular trabeculae for compaction is believed to be correlated with congenital or acquired mutations of genes associated with cardiomyocytes and the powerhouse of the cell, mitochondria. Unfortunately, complications arising from disruption in the compaction process, referred to as noncompaction, can result in heart failure and neonatal death [12].

In normal human embryo development, the compaction of myocardium begins after the 5th week of embryonic life. To form a well-developed compacted epicardial and a well-compacted endocardial layer, vascular endothelial growth factor (VEGF) and angiopoietin [13] triggers these loosely interwoven muscle fibers of the myocardium to condense, thus removing the large flattened trabeculae spongy mesh from early embryonic development prior to resolving the intertrabecular recesses into capillaries. This gradual progression of trabeculae compaction occurs faster in the LV than in the RV. However, the etiology of LVNC is poorly understood and occurs as a spectrum of pathological conditions, ranging from asymptomatic to the risk of RV failure and fetal heart failure [14]. Due to its relatively low prevalence in combination with less sensitive detection techniques for noncompaction, less is known regarding its etiology and therapy compared to other cardiomyopathies. While the occurrence of LVNC is independent of sex or age, the prevalence varies among different ethnic groups: with a greater occurrence within black populations. Lastly, the pathophysiological impact of LV noncompaction is poorly understood, though it is expected to a have an association with altered ventricular systolic and diastolic function [15, 16].

Advertisement

2. Cardiac energy metabolism: role of the mitochondria

Cardiac energy metabolism relies on regular mitochondrial function and energy production. Thus, inborn errors of metabolism impacting the mitochondrial dysfunction are a common pathological factor of LVNC. Mitochondrial cardiomyopathy is a myocardial condition defined by an aberrant heart-muscle structure or function, that involves mitochondrial respiratory chain pathways. Failure in the regulation of these metabolic pathway mechanisms can lead to cardiac dysfunction associated with non-compaction [17]. Thus, the proper functioning of mitochondrial enzyme complexes is crucial for the normal progression of cardiac morphogenesis. This review focuses specifically on the mitochondrial influence on trabecula morphogenesis and its influence on the compaction of LV myocardium. Moreover, we describe the mechanisms contributing to LVNC and explore the current diagnostic and therapeutic strategies available for this type of mitochondrial myopathy [18, 19].

A double membrane-bound organelle, mitochondria have their own genome and exhibit maternal inheritance patterns. The mitochondrial genome is composed of 22 tRNAs, 2 rRNAs, and 37 genes encoding 13 different proteins that form subunits of enzyme complexes within the oxidative phosphorylation system (OXPHOS). Although comparatively smaller than the nuclear genome, the human mitochondrial genome possesses thousands of mitochondria, each encoding dozens of copies of the mitochondrial genome. Unlike the nuclear genome, mitochondria have higher mutation rates (100 fold higher) due to the absence of a mismatch base repairing mechanism [20]. Notably, deficiencies in NADH-coenzyme Q (CoQ) reductase (complex I) and cytochrome-c oxidase (complex IV) have been commonly studied in many mitochondrial disorders [20, 21]. OXPHOS pathway involves around 80 different proteins to make five electron transport complexes (complex I–V) [22]. Many of the mutations and defects in genes involved in OXPHOS impacts the mitochondrial energy production. As such they are the primary source of reactive oxygen species (ROS) generation. Hence, the overproduction of ROS causes mitochondrial damage through a range of pathologies that affects the rest of the cell (Figure 2). In addition to oxidative phosphorylation system, mitochondria also control apoptosis and cytosol calcium concentrations. Importantly within the cardiac muscle, calcium signaling propagated within the cytosol, is trafficked through the mitochondrial membrane via the voltage dependent anion channel (VDAC) and an inner membrane located calcium uniporter or a Na+/Ca2+ exchanger. Thus, a vital feature of cellular life any alterations in mitochondrial Ca2+ homeostasis can cause profound effects in the mitochondrial function with defects contributing to cell death [23].

Figure 2.

A variety of roles played by Mitochondria are critical for cellular maintenance: Energy production (“powerhouse of the cell”), OXPHOS pathway, alters between fusion and fission cycle, Ca2+ level control and apoptosis.

Mitochondrial energy production is influenced by maternally inherited mitochondrial DNA (mtDNA) and nuclear DNA [24] with mitochondrial gene mutations causing irregular function. Energy released upon electrons transfer between the 5 enzyme complexes of the mitochondrial electron transport chain (ETC) within the inner mitochondrial membrane, acts as a proton pump across the mitochondrial membrane. This electrochemical gradient is important for cellular energetic homeostasis [25]. Defects in the five enzyme complexes of mitochondrial ETC, some of which are encoded by mitochondrial DNA, can reduce ATP energy generation resulting in an energy insufficiency. Additionally, cardiac mitochondrial function is also coupled with ROS production from oxidative damage during the OXPHOS cycle. Elevated free iron levels and defective ETC as a result of the impaired intramitochondrial metabolism is associated with a vast increase in free radical generation leading to oxidative damage that could impact development.

Cardiomyocytes are composed of bundles of myofibrils containing myofilaments. The distinct, repeating micro units of myofibrils, sarcomeres, are the fundamental contractile units of the myocytes. The mitochondria in cardiomyocytes localized to sarcomeres form intracellular energetic unit (ICEU) [26]. Catabolism favors the movement of the chemical gradient from mitochondria in the form of ATP to sarcomeres. Composed of thin and thick filaments, the thick filamentous myosin motor protein component of the cardiac sarcomere is primarily responsible for force generation. While the structural abnormalities associated with LVNC has been well described, many of the details of its mechanism have yet to be elucidated. However, studies suggest a correlation between LVNC and myocardial abnormalities with energy generation, transfer and utilization as possible contributing factors to the irregular contraction and relaxation of cardiac muscles. As such, the impact of mitochondrial related gene deletion, mitochondrial function, and energy production cannot be overstated in light of the disorders arising from disruption of these processes and their impact on compaction at the early development of the LV myocardium.

Advertisement

3. Left ventricular noncompaction (LVNC)

Left ventricular noncompaction (LVNC), also knowns as left ventricular hyper trabeculation (LVHT) or spongy myocardium, is a rare congenital myocardial dysfunction resulting from the arrest of the compaction phase during embryogenesis. Gross histology of this heterogeneous disease condition is characterized by a spongy left ventricle (LV) myocardium, abnormal prominent trabeculations at the ventricular apex, and intra-trabeculae recesses adjacent to a well compacted LV myocardium during the final stages of cardiac development [27]. Within the ventricular cavity the double-layered ventricular myocardium consisting of epicardial and endocardial layers, there appears finger-like protrusions from the myocardium with intertrabecular recesses separating trabeculations. Pathologically these anatomical features of a noncompacted heart demonstrate a tremendously thickened endocardial layer with abnormal trabeculation and a compacted thin epicardial layer. In 1984, the first case of LVNC was described in a newborn that was also associated complications of atresia of semilunar valves and intact ventricular septum [28]. Since then, of the different phenotypes of LVNC (8 different subtypes identified to date) [29], isolated LVNCs (first reported in 1926 occurring as a result of trabeculae morphogenesis interference in the absence of any cardiac abnormalities) and congenital LVNCs are the most commonly studied. With the latter type of LVNC being associated with systemic diseases including metabolic or mitochondrial diseases, cardiomyopathies such as hypertrophic, dilated, restrictive, and arrhythmogenic, congenital heart diseases, and complex syndromes affecting vast majority of the organs and tissue [30].

While rare forms of LVNC can occur from acquired mutations, commonly LVNCs are from genetic alterations transmitted through generations [31] with around 50% of the identified LVNC patients having either autosomal dominant, autosomal recessive, or X-linked genetic inheritance patterns with multiple genes modifications [32, 33, 34] (Figure 3). Predominate genetic variants associated with LV noncompaction include defects in mitochondrial, sarcomeric, and cardiac genes. In addition to mitochondrial DNA mutations [35], a wide variety of mitochondrial abnormalities significantly influence LVNC including but not limited to mitochondrial dysfunction [36], metabolic myopathy altered by mitochondrial energy production [37], dysmorphic mitochondria [31, 38], mitochondrial DNA transitions (a type of DNA mutation where a purine or pyrimidine nucleotide is replaced with its complementary purine or pyrimidines, respectively) [39, 40], deficiencies in mitochondrial respiratory chain function [41], mutations in overlapping regions [42], and mitochondrial genome mutations [43]. Further supporting mitochondrial involvement, mitochondrial numbers are enriched in the cardiac muscles of individuals with LVNC, and mitochondrial DNA (mtDNA) content and copy number are also largely altered in non-compacted hearts.

Figure 3.

Schematic overview of influencing factors in myocardial noncompaction: Differential contribution of mitochondria towards LVNC via mitochondrial dysfunction (mtDNA mutations, transitions, ETC defects, mt genome mutations) and involvement of genes associated with cardiomyopathies (sarcomeres, ion channels, cytoskeletal, chaperones, desmosomes).

Advertisement

4. Diagnostics and therapeutic aspects of LVNC

The occurrence of LVNC has increased in recent years with a prevalence of 0.01–0.3%, a mortality rate of 5–12%, and is predominately found in males [44]. While most LVNC patients are asymptomatic or noted to have mild symptoms of chest pain, later stages of LVNC can lead to sudden cardiac death due to compromised systolic function. Although the initial diagnosis can be at any stage in life, most individuals with LVNC go undiagnosed until the 5th decade of life. Symptoms are diverse in patients with LVNC and can include heart failure, cardiac arrhythmias, fatigue, excessive sweating, breathing difficulties, compromised growth, and abnormal weight gain, while others do not exhibit any symptoms [44, 45, 46]. Hence, early diagnosis is key for timely supportive care.

Common diagnostic tools available for LVNC include echocardiography (echo), cardiac magnetic resonance imaging (cardiac MRI or CMR), electrocardiogram (ECG), cardiac computed tomography (CT), coronary angiograms and myocardial biopsy based on the myocardial thickening [47]. In most cases the first-line diagnostic testing of any heart diseases is echo, due to its availability and being low-cost in nature. Echo criteria include (1) noncompacted/compacted ratio of the two-layered endocardium of >2, (2) left ventricular deep endomyocardial trabeculations, and (3) deep recesses filled with blood visualized on color Doppler [48, 49]. However, the detection of LVNC in its early stage of development are challenging, with only around 0.3% of the patients being referred for an echo [50, 51]. Additionally, echo diagnostics are sometimes considered to be a ‘too sensitive of a method’ as overdiagnosis frequently occurs, particularly in the black patient population. Therefore, serial echo diagnosis is recommended in LVNC [52].

Alternatively, CMR is a well-established, high resolution, noninvasive, albeit more expensive approach, to confirm LVNC after an initial diagnostic echo. CMR offers an enhanced sensitivity, as CMR can detect myocardial trabeculations that can be enhanced with steady-state free-precession (SSFP) cine imaging [53, 54]. In CMR, the myocardial trabeculations, apex region and end-diastole ratio can be well monitored over time. Although CMR is associated with many challenges including a wide diagnostic testing timeline, high costs, and availability, it still remains the best approach as a diagnostic tool for LVNC. Unlike echo and CMR, CT imaging is less commonly used due to the ionizing radiation and lower detection ability of myocardial tissues. Nevertheless, this tool is widely used in evaluating other disease processes such as coronary arteries. Apart from the above-mentioned diagnostic tools, advances using blood for whole exome sequencing (PGxome diagnostic exome test) and copy number variation detection can be performed to study single gene defect or defects in gene sets associated with LVNC [55].

In research settings and invasive cardiac catheterization techniques, a more invasive method of histologic examination of cardiac muscle tissue biopsies is assessed. Histological analysis can identify abnormalities in the mitochondrial appearance as ragged red fibers, indicative of mitochondrial dysfunction, are suggestive of mitochondrial anomalies (Gomori Trichrome stain, Figure 4). A typical drawback of Gomori Trichrome Staining is the absence of these ragged red fibers in children, and therefore, it fails to detect mitochondrial disorders in neonates at early stages [56, 57]. An alternative method of early detection is the examination of cardiac tissue under electron microscopy for swollen disorganized mitochondria containing irregular cristae [57].

Figure 4.

Gomori Trichrome staining of cardiac tissue from an (a) hyper trabeculated heart and a (b) healthy heart: (a) arrows indicating dysmorphic mitochondria scattered as red-ragged fibers below the matrix of the membrane, (b) mitochondria appear organized and distributed across the cardiac tissue.

Importantly, around 40% of patients identified with LVNC are determined to have a familial genetic defect that impacts only cardiac function. However, while many genetic syndromes, metabolic disorders, and mitochondrial related anomalies are also associated with LVNC, in many cases the cause of LVNC is still unknown. As there is no cure to date, routine cardiac screening is an important part of the treatment of familial LVNC, with supportive therapy for LVNC patients being dependent on individual symptoms. Supportive therapy targeted to improved quality of life and overcome cardiac dysfunction includes medications to treat heart failure such as angiotensin-converting enzyme inhibitor, beta blockers, blood thinners, diuretics, and digoxin. In severe cases of LVNC, pacemaker implantation, cardioverter defibrillator, or heart transplantation is recommended. A recent case study in 2018 by Kimura et al. [58], showed successful treatment of a near-fatal ventricular arrhythmia in an infant with LVNC using cardiac resynchronization therapy (CRT). These studies determined that with the implantation of a cardioverter defibrillator (preventive measure for cardiac arrest), dual chamber pacing initiation and CRT, effectively treated the mechanical dyssynchrony of the heart, with subsequent improvement in cardiac function.

Currently, there are no mechanisms to prevent LVNC or to return gain of cardiac function. While genetic testing can be done to identify gene mutations (Table 1) and determine the risk of passing these mutations to offspring, there are not therapeutic options to actively treat these gene mutations [74]. Future therapy that promotes personalized gene therapy could be a useful tool in treating LVNC. However, this approach could be highly challenging due to the complex genetics of LVNC.

Age/mean age at diagnosisPatient genderComplications observedDiagnostic approachReference
7 years5 males and 3 femalesSVD, VA, SE, PVC, FDHolter monitoring, intracardiac electrophysiological studiesChin et al. [59]
47 years24 males and 41 femalesHypokinesia, Akinesia, DyskinesiaEchocardiographyLofiego et al. [60]
Not reported821 males and 627 femalesLV dilatation, hypertrophy, and systolic dysfunctionTransthoracic echocardiography, MRITamborini et al. [61]
33-year-oldFemaleMyocardial sinusoids, high systolic ventricular pressure, external dyspnea, sinusoid anomaly2D-echocardiographyEngberding et al. [28]
11 years13,306 males and 12,283 femalesLower LVEF, higher LV ESVEchocardiographyBorresen et al. [51]
47.4 years183 males and 155 femalesLower LVEF, dyspnea, cardiomyopathy, left atrial enlargementTransthoracic echocardiography and or MRIVaidya et al. [12]
90-year-oldMaleAsymptomatic until heart failure followed by progressive shortness of breath, body weaknessColor-flow Doppler echocardiography, electrocardiogramCevik et al. [62]
9 years8 males and 1 femaleMostly asymptomatic, LVHElectrocardiogram, chest X-ray, 2D-echocardiogram with Doppler interrogationAlehan et al. [63]
53 years71 males and 29 femalesLVHT, associated neuromuscular disordersEchocardiographyStollberger et al. [64]
≤21 years24 males and 18 femalesMyocardial dysfunction, CVM, DCM, HCMEchocardiography, Genetic testing (cardiomyopathy gene panel)Miller et al. [65]
(2017)
62-year-oldFemaleCongestive heart failure, Bilateral crackles (mild symptoms)Echocardiography, Color Doppler imagingLin et al. [66]
6-month-oldInfant (female)Incessant ventricular fibrillation, cardiopulmonary arrest, polymorphic ventricular tachycardia, LV hypokinesisTransthoracic echocardiography, Chest X-ray, Color Doppler imagingKimura et al. [58]
46 years312 males and 2188 femalesCardiovascular and all-cause mortality, thromboembolic complications, ventricular arrhythmiasEchocardiography, MRIAung et al. [30]
41-year-oldMaleChest pain, biatrial enlargement, right ventricular hypertrophy, global hypokinesis, lower LVEFElectrocardiogram, MRITian et al. [67]
Not reported (wide range of age groups studied)6 males and 8 femalesVSD, histiocytoid cardiomyopathy, hydrops fetalis, sudden death, heart failureEchocardiographyBurke et al. [68]
29-year-oldMaleDyspnea, pulmonary rales, fatigue, cardiomegaly, hepatomegaly, splenomegalyTwelve-lead electrocardiography, twenty-four-hour Holter monitoring, speckle-tracking echocardiography, MRI, chest X-rayToader et al. [69]
57-year-oldFemaleHeart failure, onset dyspneaEchocardiographyLoria et al. [70]
36-year-oldMaleShortness of breath, exercise intolerance, orthopnea, paroxysmal nocturnal dyspneaTransesophageal echocardiogram, endomyocardial biopsy, transthoracic echocardiogramAyesha et al. [71]
69-year-oldMaleAtrial fibrillation, myocardial fibrosisElectrocardiogram, post-cardiac arrest transthoracic echocardiogramBath et al. [72]
60-year-oldFemaleIntermittent chest pain, increasing shortness of breath, decreased appetite, paroxysmal nocturnal dyspnea, orthopneaEchocardiography, chest X-ray, CT scanKalavakunta et al. [73]

Table 1.

Genetic details and characteristics identified in clinical studies.

Advertisement

5. Genes and related mutations associated with LVNC

A variety of gene mutations studied concerning other cardiomyopathies such as genes coding for sarcomere proteins [75], ion channels, cytoskeletal genes [76], chaperone proteins [77], and cellular signaling pathways have a significant association with LVNC [11, 78]. Specific genetic alterations include genes encoding β-myosin heavy chain (MYH7) [34, 79], α-cardiac actin (ACTC1) [80], and cardiac troponin T (TNNT2) [81], lamin A/C (LMNA) [82], ZASP (LDB3) [83], and taffazin (TAZ) [83, 84], cardiac myosin-binding protein C (MYBPC3) [85], and cardiac troponin I (TNNI3) [86] are associated with LVNC (Table 2).

Affected genesGene nameProtein typeDNA variantCardiac manifestationRef no
DTNAα-DystrobrevinCytoskeletal proteinC362T, N49SNLVNC, LVNC[15, 83]*
LAMP2Lysosome-associated membrane protein-2Membrane glycoproteinG928A transitionHCM, skeletal myopathy, DCM[16, 87]
TAZ (G4.5)TafazzinPhospholipid trans acylaseGIVS8-1CILVNC, DCM, BTHS[75, 83]
HCN4Hyperpolarization-activated cyclic nucleotide channel 4Ion channelG482R, Y481HBrugada syndrome, Bradycardia, LVNC[88]
SCN5ACardiac sodium channelIon channelH558RBrugada syndrome, LVNC, DCM[89]
TMEM70Transmembrane protein 70Mitochondrial membraneY112X, 578delCAHCM[15]*
Nkx2–5Nkx2-5Transcription factorA118SLVNC, DCM, HF, AVB[90]
FKBP12 (aka Fkbp1a)Peptidyl-prolyl cis-trans isomeraseImmunophilin protein familyLVNC, VSD, DCM, CHD[77, 91]
DNJAC19Mitochondrial import inner membrane translocase subunit TIM14DnaJ Hsp40 member C191G-C transversion in intron 3DCM, DCMA[16]*
BMP10Bone morphogenetic protein 10TGF-β super family proteinsV407ILVNC, VSD, HT[92]
LDB3LIM-domain binding 3Z-disk proteinG1876A, G163AILVNC, LVNC3, DCM, HCM[15, 83, 93]
MYBCP3Myosin binding protein CSarcomere thick filamentN948T, G490R, A833TDCM, LVNC10[82, 85, 94]*
MYH7β-Myosin heavy chain 7Sarcomere thick filamentL1793P, L387FLVNC5, HCM, DCM[33, 90, 94, 95, 96]*
ACTCα-cardiac actinSarcomere thin filamentA21V, E101KLVNC4[97, 98]
TNNT2Cardiac troponin T, type 2Sarcomere thin filamentR141W, R131W, E96KDCM, HCM, RCM, LVNC6, LVD[75, 81]*
TNNI3Cardiac troponin ISarcomere thin filamentsD190G, A2VRCM, DCM[86, 99]
TPM1Tropomyosin-1Actin binding proteinE40K, E54K, D159N, L113VLVNC9, DCM, HCM[16, 75, 99]*
LMNALamin A/CNuclear membrane proteinR644C, R190W, V445ELVNC[82, 100]
MT-ATP6Mitochondrially encoded ATP synthase membrane subunit 6′ATP synthase Fo subunit 6T8528C, G8529A, C8558T, A9058GLVNC[101]
MT-ATP8Mitochondrially encoded ATP synthase membrane subunit 8ATP synthase Fo subunit 8A8381G, C8858TLVNC[31, 96, 102]
MT-ND1NADH: ubiquinone oxidoreductase core subunit 1NADH dehydrogenase 1 enzymeT3398C, T4216C, G15812A, A3397GLVNC, MELAS, LHON[40, 96]
MT-TL1Mitochondrially encoded tRNA leucine 1transfer RNA (UUR)A3243G, A8344GMELAS, MERRF[16]
ANT2Peroxisomal adenine nucleotide transporter-2ADP/ATP translocase 2G1409T polymorphismDCM, LVNC/LVHT[103]
MIB1Mindbomb homolog 1E3 ubiquitin ligaseV943F, R530XLVNC7, RVNC, DCM, HF[78]
PRMD16PR/SET Domain 16Transcription regulatorK702X, N816S, P291L, L887P, V1101M, 1573dupC in exon 9LVNC8, DCM[75, 104]*
PLEC1PlectinCytoskeletal proteinG4891T, G1019AARVC, DCM, LVNC/LVHT[105]
MT-CYBCytochrome BComponent of complex IIIT15693C, A15662GLVNC, HCM[40, 96]
ACTN2α-Actinin 2Cytoskeletal proteinM228T, L727RHCM, DCM, LVNC[106]
PRKAG2Protein kinase AMP-activated non-catalytic subunit gamma 2AMP-activated protein kinase (AMP-K)H142A, T400N, N488I, E506K, R302QHCM, LVH[16, 107]
PLNPhospholambanIntegral membrane proteinR9C, L39XDCM[108, 109]

Table 2.

Genes and associated mutations in cardiomyopathies.

ILVNC, isolated left ventricular noncompaction; NLVNC, non-isolated left ventricular noncompaction; LVHT, left ventricular hyper trabeculation; RVNC, right ventricular noncompaction; AVB, Atrio ventricular block; HF, heart failure; DCM, dilated cardiomyopathy; HCM, hypertrophic cardiomyopathy; VSD, ventricular septal defect; BTHS, Barth syndrome; DCMA, dilated cardiomyopathy with ataxia syndrome; HT, hyper trabeculation; MELAS, mitochondrial encephalomyopathy; MERRF, myoclonic epilepsy with ragged-red fibers disorder; ARVC, arrhythmogenic right ventricle cardiomyopathy; CHD, congenital heart diseases; LVD, left ventricular dilation; RCM, restrictive cardiomyopathy [33].

Sarcomere gene mutations can lead to noncompaction with reduced ventricular function and hyper trabeculation [110]. Alpha-cardiac actin gene (ACTC) codes for cardiac muscle-specific alpha-actin protein, present in cardiac sarcomeres and cytoskeletal proteins, and is responsible for heart muscle contraction and generation of force to support the contraction. Novel mutations in the ACTC gene are linked to LVNC cardiomyopathies. Although, the novel protein-level amino acid sequence (A21V) mutations of ACTC1 resulting in familial LVNC are rare, it causes diverse cardiac anomalies. This resulting missense mutation in the ACTC1 gene creates structural changes to sarcomeres and their anchorage. In addition to these changes, these variants also modify the highly conserved nature of actin-like domains of the protein resulting in its destabilization with pathogenic consequences [80]. The MYH7 gene encoding for myosin heavy chain beta (MyHC-β) isoform is another primary sarcomeric protein in the adult heart. The ATPase activity of MYH7 powers the myosin power stroke within the myosin heads to convert energy that propels shortening of the sarcomeres. A heterozygous missense mutation (I467T) in MYH7 found in inherited cardiomyopathies presenting with a high penetrance and sudden death, can result in LVNC and hypertrophic cardiomyopathy (HCM) [111]. As the mutation site in MyH7 is close to the hydrophobic ATP binding pocket of the motor domain, the amino acid replacement of hydrophobic isoleucine with a non-polar threonine alters the structure subsequently effecting the ATPase activity of the motor domain [79, 80]. Cardiac Myosin binding protein C (MYBPC3) is another LVNC linked sarcomere protein. Infants with pathogenic truncating mutations in MYBPC3 die at birth from HCM and LVNC complications. These mutations are believed to cause alterations in the primary contractile function of the heart and septal defects. However, the absolute relationship of contractile dysfunction to sarcomere protein mutations in the progression and pathophysiology of LVNC remains poorly understood [112].

Notch signaling is a prime mediator of cardiac embryogenesis. Despite the anomalies in other signaling pathways, the dysfunction of the Notch pathway has a notable contribution to LVNC. Germline mutations in human myocardial MIB1 (mind bomb homolog 1) coding for E3 ubiquitin ligase that promotes the endocytosis of Delta and Jagged, (NOTCH ligands) are involved in LV noncompaction [113]. In these studies, MIB1 protein was noted to be vital in activating the NOTCH signaling pathway while cardiac deformity managed by two of the MIB1 autosomal dominant mutations (V943F and R530X) causes deregulation of Notch signaling resulting in LVNC. Furthermore, Luxan et al. [78] reported that myocardial inactivation of MIB1 results in the enlargement of the ventricular trabeculae and a thin compact myocardium.

Alterations in cytoskeletal genes have been linked with the LVNC development. The intermediate filament protein, lamin (lamin A/C) or LMNA, maintains the mechanical integrity of the nuclear envelope. Heterozygous LMNA gene mutations (R644C and R190W) are linked to familial and sporadic LVNC, although the mechanisms have not been elucidated to date. Furthermore, lamin A/C V445E mutations were found to be related to sudden death in LVNC patients [82]. Cardiac expressing alpha-dystrobrevin (DTNA) belongs to a dystrophin-related protein family that is significant in intracellular signal transduction. Genetic alterations in DTNA and dystrophin-associated glycoprotein complex (DAPC) located in sarcolemma provoke myocardial disorders targeting the systolic function of the heart [41]. Inactivation of DTNA leads to muscular dystrophy, skeletal myopathy, and cardiomyopathy characterized by deep trabeculations which are always observed in LVNC [76]. Congenital LVNC frequently accompanies neuromuscular disorders in patients with myotonic dystrophy type 1 (MD1), an autosomal dominant type of disorder resulting from a trinucleotide expansion (CTG nucleotide repetitions) in the dystrophia myotonica protein kinase (DMPK) gene [114].

Advertisement

6. Differential contribution of mitochondria towards LVNC

6.1 Mitochondrial DNA (mtDNA) mutations leading to dysmorphic mitochondria

mtDNA sequence variants are maternally inherited point mutations representing a predominate cause of inherited diseases within a tissue or organ. Studies performed on mitochondria isolated from patient’s blood and in vivo cardiac tissues acknowledged the contribution of mtDNA mutations in the formation of cardiac noncompaction [115, 116]. In these studies, instability of cardiac-specific mtDNA is coupled to metabolic irregularities that impact cardiac functionality in response to a myriad of mitochondrial dysfunctions. Liu et al. [116] determined that hearts with noncompaction present with mitochondrial structural anomalies and have a reduced mtDNA copy number with the G4.5 gene on Xq28 that codes for acyltransferase tafazzin (TAZ); the first gene to be studied in detail in relation to the etiology of LVNC [117]. Tafazzin is a mitochondrial membrane component vital for proper functioning of the electron transport chain. This X-linked TAZ gene encodes for the mitochondrial membrane-associated phospholipid-modifying enzyme that alters cardiolipin through the addition of linoleic acid. As cardiolipin is a major player in the mitochondrial inner membrane and is requisite for the maintenance of mitochondrial shape, protein transport, and energy production within cells, it is essential for maintaining the organization of mitochondrial cristae [57]. TAZ gene mutations resulting in Bath Syndrome, perturbed mitochondrial cardiolipin metabolism and are associated with dilated cardiomyopathy. Inactivation of the TAZ gene produces dysfunctional tafazzin proteins. In this circumstance, linoleic acid is not present to alter cardiolipin. As a result, problems with normal mitochondrial shape and functions such as energy production and protein transport develop. High energy-demanding tissues including heart are more susceptible to cell death due to decreased mitochondrial energy production [118]. Furthermore, TAZ deficiency impairs cardiolipin remodeling and alters the assembly and stability of mitochondrial super complexes of complex III and IV (intermittently impacting complex V as well) in the inner mitochondrial membrane [119].

These effects occasionally result in respiratory chain activity malfunctions. Mitochondrial transcription factor A (TFAM) is a nuclear-encoded protein that regulates the transcription, packaging, and stability of the mitochondrial genome. TFAM is crucial for the development and differentiation of the mitochondrial genome [120]. It is highly likely that TFAM regulates the mtDNA copy number by binding to the mtDNA promoter region to initiate transcription. Thus, the impact of TFAM on mtDNA copy number illustrates the direct influence of TFAM in mitochondrial function. Carnitine palmitoyl transferases (CPT) are mitochondrial enzymes involved in the transportation of Acyl-CoA to the mitochondrial Matrix for β-oxidation. Located in the outer mitochondrial membrane, CPT I convert fatty acyl CoA into fatty acylcarnitine. Located in the inner mitochondrial membrane, CPT II reconverts fatty acylcarnitine into free carnitine and fatty acyl CoA for β-oxidation for energy generation. Inherited defects in CPT I, II or CPT translocases can result in failure of long-chain fatty acids transportation into the mitochondria for oxidation. This can result in a significant reduction in the ratio of free CoA to acyl CoA thus ultimately disrupting metabolic pathways and decreasing cardiac ATP production [24].

6.2 Mitochondrial dynamics and dysfunction

Adult cardiomyocytes are enriched with hypo-dynamic mitochondria that lack an interconnected reticular network. The fusion-fission cycle of cardiac mitochondria is common in cardiomyocyte mitochondrial dynamics [121]. Regulated by specialized proteins, enzymes, and adapter proteins that physically alter mitochondrial membranes, a delicate balance between mitochondrial fusion and mitochondrial fission is important in the regulation of cellular health. In adult cardiomyocytes progressive mitochondrial fission and fusion cycles occur frequently with the aid of proteins needed for the mitochondrial network remodeling. The key aspects of mitochondrial dynamics can be studied with cardiac-specific ablation of mitochondrial fission and fusion protein genes. As the cardiomyocyte mitochondria exhibit a certain degree of dynamism [122, 123], inability for the fusion or fission process could result in smaller or enlarged mitochondria, respectively. Hence, the imbalance between constant ongoing process of fusion and fission cycle are associated with increased caspases activity and subsequent cell death.

Mitochondrial dysfunction is frequently interrelated with LV noncompaction. Primary mitochondrial dysfunction arises from mutations in mitochondrial proteins involved with OXPHOS. Mitochondrial dysfunction refines mitochondrial dynamics with this phenomenon being associated with a variety of cardiac pathologies. For example, the MYH7 coding myosin protein is associated with the structure and function of cardiac sarcomere. Besides, the mutations in MYH7 linked with HCM and LVNC, makes myosin an important protein in cardiac function as defects in MYH7 expression are a common cause of cardiomyopathy [79]. Friedreich’s ataxia (FRDA) is an example of a heart disease arising from mitochondrial dysfunction. FRDA is caused by the trinucleotide GAA expansion in the first intron of FXN gene (codes for mitochondrial matrix iron chaperone protein, frataxin) [123] with this triplet expansion resulting in transcription silencing and diminished frataxin expression. As a result, this mitochondrial localized protein influences iron homeostasis and respiratory control. Therefore, impaired FXN gene function in FRDA patients is associated with dysfunctional ETC complexes, specifically the complex III with high ROS levels suggesting altered mitochondrial function [124].

6.3 Mitochondrial metabolic myopathy resulting from respiratory chain complex deficiency

Mitochondrial cardiomyopathy, a distinct myocardial condition is denoted by improper cardiac muscle structure and function accompanied by genetic defects in the mitochondrial respiratory chain. Various genetic anomalies are detected in genes regulating mitochondrial activities or in genes coding for ETC complexes or in genes that code for proteins necessary in assembling and transporting.

Peroxisomal adenine nucleotide transporter-1 and 2 (ANT-1 and ANT-2) in humans are ADP/ATP carriers with two main functions; to catalyze mitochondrial-cytosol ATP/ADP exchange and regulation of the mitochondrial inner membrane mitochondrial permeability transition pore (mtPTP). The ATN-1 isoform influencing the mtPTP towards a more open state while ATN-2 is involved in the guiding the closure of the mtPTP. Within the heart, muscles, and brain ANT-1 is the predominant isoform while the second isoform ANT-2 is expressed systemically. However, ablation of ANT-1 isoform does not impair fetal development; but does result in hypertrophic cardiomyopathy in the postnatal period. In contrast, a study published in 2016 by Kokoszka et al. [125] identified the embryonic lethality of ANT-2 null mutations to be associated with a distinct cardiac developmental defect consistent with LVNC/LVHT; characterized by swollen mitochondria in cardiomyocytes and hyperproliferating cardiomyocytes. In these studies, mice with ANT-2 mutations, cardiac maturation progresses during embryonic days E9.5 and E13.5. However, by day E14.5 mice exhibit embryonic lethality. Histologic analysis of the heart revealed hyper-proliferating cardiomyocytes, decreased apoptosis, and swollen mitochondria with few cristae, and mtPTP open for transport. These findings suggest that loss of ANT-2 regulation of mtPTP closure results in cardiomyocytes maintenance in an immature state with a reduction in total number of contractile fibers and loss of organization thus paving the way for the continued immature cardiomyocyte proliferation resulting in LVNC and embryonic lethality [37].

6.4 Mitochondrial genome mutations in LVNC

In most of the reported cases, LVNC is related to the alterations in nuclear DNA encoded mitochondrial genes. Defects in the mediator of a mitochondrial fusion protein, Mitofusins 1 & 2 (MFN 1 & 2) affect the mitochondrial morphology during embryogenesis. Mediated by MFN1 and 2, mitochondrial fusion facilitates the exchange between the contents of mitochondrial membrane and matrix to help in maintaining the mitochondrial function. Mutations in any of the MFN’s results in mitochondrial fragmentation that affects the symmetry of fusion and fission events [126]. As cell apoptosis is significantly influenced by the mitochondrial fusion and fission, cells lacking apoptosis related genes such as caspases 3, 7, 8, FADD, and c-FLIP are also susceptible to develop cardiac noncompaction. Caspases 3 and 7 are potential regulators of mitochondrial apoptosis. A3243G transition in tRNALeu(UUR) gene is another common mitochondrial mutation [127, 128].

The tRNALeu(UUR) gene encodes for mitochondrial transfer ribonucleic acid (tRNA) for leucine (tRNALeu) and this nucleotide alteration was studied in both HCM and LVNC [129]. Other studies show that A3243G mutation related disorders cause alterations in nearly 56 different genes, thus having a broad spectrum of impact. The A to G transition at the 3243rd position in tRNALeu (UUR) gene results in diminished glucose oxidation rate, NADH response, and mitochondrial membrane potential that ultimately affects ATP production [127, 130]. Lastly, a A8381G transition in ATPase subunit 8 genes (ATPase 8) was observed in a patient having LVNC complications [43, 102, 115]. These arising missense mutations impact within the ETC complex V that could alter the ATPase complex stability, directing to reduced ATP production [43, 131].

6.5 Mitochondrial DNA transitions in LVNC

Single mutations arising from mtDNA transitions are responsible for an abnormal diseased phenotype. mtDNA transitions mutations occur when there is a replacement of the complementary nucleotide (A replaced with G, T replaced with C and vice versa). One example is found in the mitochondrially encoded NADH: ubiquinone oxidoreductase core subunit 1 (mt-ND1) encoding gene for the NADH dehydrogenase 1 enzyme that is part of complex ETC complex I, which is active in mitochondria. Here, a single nucleotide transition (T → C) at a highly conserved nucleotide position (T3398C) is within the region of the ND1 gene of the mt genome. This particular mtDNA transition results in the conversion of methionine to threonine at the 31st amino acid position was identified in patients suffering from cardiomyopathy [39]. A case report presented by Finsterer et al. [40] investigated the combination of A → G transition at the nucleotide position of 15,662 in novel mitochondrial cytochrome B and three of the known mutations (T3398C, T4216C, G15812A) in ND1 gene. These above-mentioned mutations were found to be in association with left ventricular hyper trabeculation and some clinical conditions. Later studies determined that a homoplasmic A → G transition at the nucleotide position of 8381 in the ATPase8 gene, investigated by Finsterer et al. [129] resulted in an amino acid substitution of alanine to threonine. Their findings revealed the correlation of LVNC with A8381G mtDNA transition that develop later in adulthood.

6.6 Mutations in overlapping regions in LVNC

The ETC complex V, known as ATP synthase, is made up of 13 protein subunits. Two of the gene mutations in mitochondrial genome or the nuclear genes result in complex V deficiency. The mitochondrial-encoded protein subunits of complex V, ATPase 6, and ATPase 8 overlap in the mitochondrial genome and undergo polycistronic fashion of transcription. The nucleotide position 8528 is within the overlapping region that codes for ATPase 6 and ATPase 8 subunits. The phenotypic representation of point mutation affects ATPase 6 and ATPase 8 genes of complex V. This is commonly seen in identical mitochondrial mtDNA mutations in children [42]. Here, the nucleotide alteration of m. T8528C, causes a M1T in the start codon in ATPase 6 subunit (One might expect to have alterations in the protein translation initiation. However, it has not been reported so far, thus making the function of altered ATPase 6 unclear) and W55R missense mutation in ATPase 8 subunit. Additionally, 2 other overlapping mutations in G8529A and C8558T affecting both the subunits have been reported in patients exhibiting cardiomyopathies [101, 115, 132]. This kind of overlapping pathogenic mutations could cause reduced ATP synthesis by affecting mitochondrial energy production and complex I, II and III for which the mechanism is still unknown [42].

Advertisement

7. Conclusion

This review overlays the aspects of gene mutations relevant to mitochondrial energetics, cardiomyocyte function, and LV myocardial noncompaction. Understanding the rudimentary causes of LVNC imparts helpful information in the prognostic evaluation and early diagnosis of LV noncompaction. Further examination of mitochondrial involvement in cardiac noncompaction leading to heart failure would facilitate advances in the therapeutic treatment of this disease process.

References

  1. 1. Grant RP. The embryology of ventricular flow pathways in man. Circulation. 1962;25:756-779
  2. 2. Krishnan A, Samtani R, Dhanantwari P, Lee E, Yamada S, Shiota K, et al. A detailed comparison of mouse and human cardiac development. Pediatric Research. 2014;76(6):500-507
  3. 3. Dhanantwari P, Lee E, Krishnan A, Samtani R, Yamada S, Anderson S, et al. Human cardiac development in the first trimester: A high-resolution magnetic resonance imaging and episcopic fluorescence image capture atlas. Circulation. 2009;120(4):343-351
  4. 4. Tan CMJ, Lewandowski AJ. The transitional heart: From early embryonic and fetal development to neonatal life. Fetal Diagnosis and Therapy. 2020;47(5):373-386
  5. 5. Zhang W, Chen H, Qu X, Chang CP, Shou W. Molecular mechanism of ventricular trabeculation/compaction and the pathogenesis of the left ventricular noncompaction cardiomyopathy (LVNC). American Journal of Medical Genetics. Part C, Seminars in Medical Genetics. 2013;163C(3):144-156
  6. 6. Sedmera D, Pexieder T, Hu N, Clark EB. A quantitative study of the ventricular myoarchitecture in the stage 21-29 chick embryo following decreased loading. European Journal of Morphology. 1998;36(2):105-119
  7. 7. Samsa LA, Yang B, Liu J. Embryonic cardiac chamber maturation: Trabeculation, conduction, and cardiomyocyte proliferation. American Journal of Medical Genetics. Part C, Seminars in Medical Genetics. 2013;163C(3):157-168
  8. 8. Keren A, Billingham ME, Popp RL. Echocardiographic recognition and implications of ventricular hypertrophic trabeculations and aberrant bands. Circulation. 1984;70(5):836-842
  9. 9. Ho SY, Nihoyannopoulos P. Anatomy, echocardiography, and normal right ventricular dimensions. Heart. 2006;92(Suppl 1):i2-i13
  10. 10. Kruithof BP, Kruithof-De-Julio M, Poelmann RE, Gittenberger-De-Groot AC, Gaussin V, Goumans MJ. Remodeling of the myocardium in early trabeculation and cardiac valve formation; a role for TGFbeta2. The International Journal of Developmental Biology. 2013;57(11-12):853-863
  11. 11. Wengrofsky P, Armenia C, Oleszak F, Kupferstein E, Rednam C, Mitre CA, et al. Left ventricular trabeculation and noncompaction cardiomyopathy: A review. EC Clinical and Experimental Anatomy. 2019;2(6):267-283
  12. 12. Vaidya VR, Lyle M, Miranda WR, Farwati M, Isath A, Patlolla SH, et al. Long-term survival of patients with left ventricular noncompaction. Journal of the American Heart Association. 2021;10(2):e015563
  13. 13. Goud A, Padmanabhan S. A rare form of cardiomyopathy: Left ventricular non-compaction cardiomyopathy. Journal of Community Hospital Internal Medicine Perspectives. 2016;6(1):29888
  14. 14. Stollberger C, Finsterer J, Blazek G. Left ventricular hypertrabeculation/noncompaction and association with additional cardiac abnormalities and neuromuscular disorders. The American Journal of Cardiology. 2002;90(8):899-902
  15. 15. Towbin JA. Left ventricular noncompaction: A new form of heart failure. Heart Failure Clinics. 2010;6(4):453-469 viii
  16. 16. Towbin JA, Jefferies JL. Cardiomyopathies due to left ventricular noncompaction, mitochondrial and storage diseases, and inborn errors of metabolism. Circulation Research. 2017;121(7):838-854
  17. 17. St-Pierre G, Steinberg C, Dubois M, Senechal M. What the cardiologist should know about mitochondrial cardiomyopathy? The Canadian Journal of Cardiology. 2019;35(2):221-224
  18. 18. Greenwell AA, Gopal K, Ussher JR. Myocardial energy metabolism in non-ischemic cardiomyopathy. Frontiers in Physiology. 2020;11:570421
  19. 19. Guclu A, Knaapen P, Harms HJ, Parbhudayal RY, Michels M, Lammertsma AA, et al. Disease stage-dependent changes in cardiac contractile performance and oxygen utilization underlie reduced myocardial efficiency in human inherited hypertrophic cardiomyopathy. Circulation: Cardiovascular Imaging. May 2017;10(5):e005604. DOI: 10.1161/CIRCIMAGING.116.005604
  20. 20. Taanman JW. The mitochondrial genome: Structure, transcription, translation and replication. Biochimica et Biophysica Acta. 1999;1410(2):103-123
  21. 21. Saada A, Bar-Meir M, Belaiche C, Miller C, Elpeleg O. Evaluation of enzymatic assays and compounds affecting ATP production in mitochondrial respiratory chain complex I deficiency. Analytical Biochemistry. 2004;335(1):66-72
  22. 22. Tuppen HA, Blakely EL, Turnbull DM, Taylor RW. Mitochondrial DNA mutations and human disease. Biochimica et Biophysica Acta. 2010;1797(2):113-128
  23. 23. Giorgi C, Agnoletto C, Bononi A, Bonora M, De Marchi E, Marchi S, et al. Mitochondrial calcium homeostasis as potential target for mitochondrial medicine. Mitochondrion. 2012;12(1):77-85
  24. 24. Winter SC, Buist NR. Cardiomyopathy in childhood, mitochondrial dysfunction, and the role of L-carnitine. American Heart Journal. 2000;139(2 Pt 3):S63-S69
  25. 25. McCormick EM, Muraresku CC, Falk MJ. Mitochondrial genomics: A complex field now coming of age. Current Genetic Medicine Reports. 2018;6(2):52-61
  26. 26. Seppet EK, Eimre M, Anmann T, Seppet E, Peet N, Kaambre T, et al. Intracellular energetic units in healthy and diseased hearts. Experimental and Clinical Cardiology. 2005;10(3):173-183
  27. 27. Reynen K, Bachmann K, Singer H. Spongy myocardium. Cardiology. 1997;88(6):601-602
  28. 28. Engberding R, Bender F. Identification of a rare congenital anomaly of the myocardium by two-dimensional echocardiography: Persistence of isolated myocardial sinusoids. The American Journal of Cardiology. 1984;53(11):1733-1734
  29. 29. Lipshultz SE, Law YM, Asante-Korang A, Austin ED, Dipchand AI, Everitt MD, et al. Cardiomyopathy in children: Classification and diagnosis: A scientific statement from the American Heart Association. Circulation. 2019;140(1):e9-e68
  30. 30. Aung N, Doimo S, Ricci F, Sanghvi MM, Pedrosa C, Woodbridge SP, et al. Prognostic significance of left ventricular noncompaction: Systematic review and meta-analysis of observational studies. Circulation: Cardiovascular Imaging. Jan 2020;13(1):e009712. DOI: 10.1161/CIRCIMAGING.119.009712
  31. 31. Finsterer J, Stollberger C, Schubert B. Acquired left ventricular hypertrabeculation/noncompaction in mitochondriopathy. Cardiology. 2004;102(4):228-230
  32. 32. Franz WM, Muller OJ, Katus HA. Cardiomyopathies: From genetics to the prospect of treatment. Lancet. 2001;358(9293):1627-1637
  33. 33. Abbasi Y, Jabbari J, Jabbari R, Yang RQ , Risgaard B, Kober L, et al. The pathogenicity of genetic variants previously associated with left ventricular non-compaction. Molecular Genetics & Genomic Medicine. 2016;4(2):135-142
  34. 34. Mazzarotto F, Hawley MH, Beltrami M, Beekman L, de Marvao A, McGurk KA, et al. Systematic large-scale assessment of the genetic architecture of left ventricular noncompaction reveals diverse etiologies. Genetics in Medicine. 2021;23(5):856-864
  35. 35. Finsterer J, Harbo HF, Baets J, Van Broeckhoven C, Di Donato S, Fontaine B, et al. EFNS guidelines on the molecular diagnosis of mitochondrial disorders. European Journal of Neurology. 2009;16(12):1255-1264
  36. 36. Scaglia F, Towbin JA, Craigen WJ, Belmont JW, Smith EO, Neish SR, et al. Clinical spectrum, morbidity, and mortality in 113 pediatric patients with mitochondrial disease. Pediatrics. 2004;114(4):925-931
  37. 37. Kokoszka JE, Waymire KG, Flierl A, Sweeney KM, Angelin A, MacGregor GR, et al. Deficiency in the mouse mitochondrial adenine nucleotide translocator isoform 2 gene is associated with cardiac noncompaction. Biochimica et Biophysica Acta. 2016;1857(8):1203-1212
  38. 38. Finsterer J, Obermann I, Milvay E. Diagnostic yield of the lactate stress test in 160 patients with suspected respiratory chain disorder. Metabolic Brain Disease. 2000;15(3):163-171
  39. 39. Jaksch M, Hofmann S, Kaufhold P, Obermaier-Kusser B, Zierz S, Gerbitz KD. A novel combination of mitochondrial tRNA and ND1 gene mutations in a syndrome with MELAS, cardiomyopathy, and diabetes mellitus. Human Mutation. 1996;7(4):358-360
  40. 40. Finsterer J, Bittner R, Bodingbauer M, Eichberger H, Stollberger C, Blazek G. Complex mitochondriopathy associated with 4 mtDNA transitions. European Neurology. 2000;44(1):37-41
  41. 41. Pignatelli RH, McMahon CJ, Dreyer WJ, Denfield SW, Price J, Belmont JW, et al. Clinical characterization of left ventricular noncompaction in children: A relatively common form of cardiomyopathy. Circulation. 2003;108(21):2672-2678
  42. 42. Ware SM, El-Hassan N, Kahler SG, Zhang Q , Ma YW, Miller E, et al. Infantile cardiomyopathy caused by a mutation in the overlapping region of mitochondrial ATPase 6 and 8 genes. Journal of Medical Genetics. 2009;46(5):308-314
  43. 43. Perucca-Lostanlen D, Narbonne H, Hernandez JB, Staccini P, Saunieres A, Paquis-Flucklinger V, et al. Mitochondrial DNA variations in patients with maternally inherited diabetes and deafness syndrome. Biochemical and Biophysical Research Communications. 2000;277(3):771-775
  44. 44. Udeoji DU, Philip KJ, Morrissey RP, Phan A, Schwarz ER. Left ventricular noncompaction cardiomyopathy: Updated review. Therapeutic Advances in Cardiovascular Disease. 2013;7(5):260-273
  45. 45. Morales-Arraez D, Ventura-Cots M, Altamirano J, Abraldes JG, Cruz-Lemini M, Thursz MR, et al. Correction to: The MELD score is superior to the Maddrey discriminant function score to predict short-term mortality in alcohol-associated hepatitis: A global study. American Journal of Gastroenterology. 2022;117(5):818
  46. 46. Cincinnati Children's. Available from: https://www.cincinnatichildrens.org/service/c/cardiomyopathy/types/left-ventricular-non-compaction-cardiomyopathy [Accessed: 12 August]
  47. 47. Bennett CE, Freudenberger R. The current approach to diagnosis and management of left ventricular noncompaction cardiomyopathy: Review of the literature. Cardiology Research and Practice. 2016;2016:5172308
  48. 48. Jenni R, Oechslin E, Schneider J, Attenhofer Jost C, Kaufmann PA. Echocardiographic and pathoanatomical characteristics of isolated left ventricular non-compaction: A step towards classification as a distinct cardiomyopathy. Heart. 2001;86(6):666-671
  49. 49. Tian T, Liu Y, Gao L, Wang J, Sun K, Zou Y, et al. Isolated left ventricular noncompaction: Clinical profile and prognosis in 106 adult patients. Heart and Vessels. 2014;29(5):645-652
  50. 50. Agarwal A, Khandheria BK, Paterick TE, Treiber SC, Bush M, Tajik AJ. Left ventricular noncompaction in patients with bicuspid aortic valve. Journal of the American Society of Echocardiography. 2013;26(11):1306-1313
  51. 51. Borresen MF, Blixenkrone-Moller E, Kock TO, Sillesen AS, Vogg ROB, Pihl CA, et al. Prevalence of left ventricular noncompaction in newborns. Circulation: Cardiovascular Imaging. 2022;15(6):e014159
  52. 52. Ross SB, Barratt A, Semsarian C. Time to reconsider the diagnosis of “left ventricular noncompaction” in adults? Heart, Lung & Circulation. 2022;31(3):301-303
  53. 53. Gleeson TG, Mwangi I, Horgan SJ, Cradock A, Fitzpatrick P, Murray JG. Steady-state free-precession (SSFP) cine MRI in distinguishing normal and bicuspid aortic valves. Journal of Magnetic Resonance Imaging. 2008;28(4):873-878
  54. 54. Paluszkiewicz J, Milting H, Kaluzna-Oleksy M, Pyda M, Janus M, Korperich H, et al. Left ventricular non-compaction cardiomyopathy-still more questions than answers. Journal of Clinical Medicine. 16 Jul 2022;11(14):4135. DOI: 10.3390/jcm11144135
  55. 55. Prevention Genetics. Available from: https://www.preventiongenetics.com/testInfo?val=Left-Ventricular-Noncompaction-%28LVNC%29-Panel [Accessed August 12]
  56. 56. Jackson MJ, Schaefer JA, Johnson MA, Morris AA, Turnbull DM, Bindoff LA. Presentation and clinical investigation of mitochondrial respiratory chain disease. A study of 51 patients. Brain. 1995;118(Pt 2):339-357
  57. 57. Meyers DE, Basha HI, Koenig MK. Mitochondrial cardiomyopathy: Pathophysiology, diagnosis, and management. Texas Heart Institute Journal. 2013;40(4):385-394
  58. 58. Kimura M, Kawano K, Yaoita H, Kure S. Successful treatment of an infant with left ventricular noncompaction presenting with fatal ventricular arrhythmia treated with cardiac resynchronization therapy and an implantable cardioverter defibrillator. Case Reports in Cardiology. 2018;2018:1-5
  59. 59. Chin TK, Perloff JK, Williams RG, Jue K, Mohrmann R. Isolated noncompaction of left ventricular myocardium. A study of eight cases. Circulation. 1990;82(2):507-513
  60. 60. Lofiego C, Biagini E, Ferlito M, Pasquale F, Rocchi G, Perugini E, et al. Paradoxical contributions of non-compacted and compacted segments to global left ventricular dysfunction in isolated left ventricular noncompaction. The American Journal of Cardiology. 2006;97(5):738-741
  61. 61. Tamborini G, Pepi M, Celeste F, Muratori M, Susini F, Maltagliati A, et al. Incidence and characteristics of left ventricular false tendons and trabeculations in the normal and pathologic heart by second harmonic echocardiography. Journal of the American Society of Echocardiography. 2004;17(4):367-374
  62. 62. Cevik C, Stainback RF. Isolated left ventricular noncompaction in a 90-year-old man. Texas Heart Institute Journal. 2012;39(2):255-257
  63. 63. Alehan D. Clinical features of isolated left ventricular noncompaction in children. International Journal of Cardiology. 2004;97(2):233-237
  64. 64. Stollberger C, Blazek G, Winkler-Dworak M, Finsterer J. Sex differences in left ventricular noncompaction in patients with and without neuromuscular disorders. Revista Española de Cardiología. 2008;61(2):130-136
  65. 65. Miller EM, Hinton RB, Czosek R, Lorts A, Parrott A, Shikany AR, et al. Genetic testing in pediatric left ventricular noncompaction. Circulation: Cardiovascular Genetics. Dec 2017;10(6):e001735. DOI: 10.1161/CIRCGENETICS.117.001735
  66. 66. Lin YN, Wang YQ , Yu Y, Cao Q , Wang F, Chen SY. Left ventricular noncompaction cardiomyopathy: A case report and literature review. International Journal of Clinical and Experimental Medicine. 2014;7(12):5130-5133
  67. 67. Tian J, Uddin A, Akhrass P. Left ventricular noncompaction: A rare case of nonischemic cardiomyopathy. Case Reports in Cardiology. 2019;2019:5637638
  68. 68. Burke A, Mont E, Kutys R, Virmani R. Left ventricular noncompaction: A pathological study of 14 cases. Human Pathology. 2005;36(4):403-411
  69. 69. Toader D, Paraschiv A, Tudorascu P, Tudorascu D, Bataiosu C, Balseanu A. Left ventricular noncompaction-a rare cause of triad: Heart failure, ventricular arrhythmias, and systemic embolic events: A case report. Journal of Medical Case Reports. 2021;15(1):316
  70. 70. Loria V, Colizzi C, Vaccarella M, Franceschi F, Aspromonte N. Left ventricular noncompaction: Cause or consequence of myocardial disease? A case report and literature review. Cardiology. 2019;143(3-4):100-104
  71. 71. Ayesha B, Ahmed R, Gomceli U, Manrique C, Nicu M, Chilimuri S. A case of isolated left ventricular non-compaction cardiomyopathy in a HIV patient presenting with acute heart failure. Cardiology Research. 2019;10(4):236-240
  72. 72. Bath AS, Aggarwal S, Gupta V, Kalavakunta JK. A case of left ventricular noncompaction presenting as atrial fibrillation. Cureus. 2019;11(3):e4309
  73. 73. Kalavakunta JK, Tokala H, Gosavi A, Gupta V. Left ventricular noncompaction and myocardial fibrosis: A case report. International Archives of Medicine. 2010;3:20
  74. 74. Shieh JT. Implications of genetic testing in noncompaction/hypertrabeculation. American Journal of Medical Genetics. Part C, Seminars in Medical Genetics. 2013;163C(3):206-211
  75. 75. Klaassen S, Probst S, Oechslin E, Gerull B, Krings G, Schuler P, et al. Mutations in sarcomere protein genes in left ventricular noncompaction. Circulation. 2008;117(22):2893-2901
  76. 76. Ichida F, Tsubata S, Bowles KR, Haneda N, Uese K, Miyawaki T, et al. Novel gene mutations in patients with left ventricular noncompaction or Barth syndrome. Circulation. 2001;103(9):1256-1263
  77. 77. Shou W, Aghdasi B, Armstrong DL, Guo Q , Bao S, Charng MJ, et al. Cardiac defects and altered ryanodine receptor function in mice lacking FKBP12. Nature. 1998;391(6666):489-492
  78. 78. Luxan G, Casanova JC, Martinez-Poveda B, Prados B, D'Amato G, MacGrogan D, et al. Mutations in the NOTCH pathway regulator MIB1 cause left ventricular noncompaction cardiomyopathy. Nature Medicine. 2013;19(2):193-201
  79. 79. Hoedemaekers YM, Cohen-Overbeek TE, Frohn-Mulder IM, Dooijes D, Majoor-Krakauer DF. Prenatal ultrasound diagnosis of MYH7 non-compaction cardiomyopathy. Ultrasound in Obstetrics & Gynecology. 2013;41(3):336-339
  80. 80. Frustaci A, De Luca A, Guida V, Biagini T, Mazza T, Gaudio C, et al. Novel alpha-actin gene mutation p.(Ala21Val) causing familial hypertrophic cardiomyopathy, myocardial noncompaction, and transmural crypts. Clinical-pathologic correlation. Journal of the American Heart Association. 10 Feb 2018;7(4):e008068. DOI: 10.1161/JAHA.117.008068
  81. 81. Luedde M, Ehlermann P, Weichenhan D, Will R, Zeller R, Rupp S, et al. Severe familial left ventricular non-compaction cardiomyopathy due to a novel troponin T (TNNT2) mutation. Cardiovascular Research. 2010;86(3):452-460
  82. 82. Liu Z, Shan H, Huang J, Li N, Hou C, Pu J. A novel lamin A/C gene missense mutation (445 V > E) in immunoglobulin-like fold associated with left ventricular non-compaction. Europace. 2016;18(4):617-622
  83. 83. Xing Y, Ichida F, Matsuoka T, Isobe T, Ikemoto Y, Higaki T, et al. Genetic analysis in patients with left ventricular noncompaction and evidence for genetic heterogeneity. Molecular Genetics and Metabolism. 2006;88(1):71-77
  84. 84. Ma L, Vaz FM, Gu Z, Wanders RJ, Greenberg ML. The human TAZ gene complements mitochondrial dysfunction in the yeast taz1Delta mutant. Implications for Barth syndrome. The Journal of Biological Chemistry. 2004;279(43):44394-44399
  85. 85. Van Driest SL, Vasile VC, Ommen SR, Will ML, Tajik AJ, Gersh BJ, et al. Myosin binding protein C mutations and compound heterozygosity in hypertrophic cardiomyopathy. Journal of the American College of Cardiology. 2004;44(9):1903-1910
  86. 86. Kimura A, Harada H, Park JE, Nishi H, Satoh M, Takahashi M, et al. Mutations in the cardiac troponin I gene associated with hypertrophic cardiomyopathy. Nature Genetics. 1997;16(4):379-382
  87. 87. Nishino I, Fu J, Tanji K, Yamada T, Shimojo S, Koori T, et al. Primary LAMP-2 deficiency causes X-linked vacuolar cardiomyopathy and myopathy (Danon disease). Nature. 2000;406(6798):906-910
  88. 88. Milano A, Vermeer AM, Lodder EM, Barc J, Verkerk AO, Postma AV, et al. HCN4 mutations in multiple families with bradycardia and left ventricular noncompaction cardiomyopathy. Journal of the American College of Cardiology. 2014;64(8):745-756
  89. 89. Shan L, Makita N, Xing Y, Watanabe S, Futatani T, Ye F, et al. SCN5A variants in Japanese patients with left ventricular noncompaction and arrhythmia. Molecular Genetics and Metabolism. 2008;93(4):468-474
  90. 90. Gifford CA, Ranade SS, Samarakoon R, Salunga HT, de Soysa TY, Huang Y, et al. Oligogenic inheritance of a human heart disease involving a genetic modifier. Science. 2019;364(6443):865-870
  91. 91. Chen H, Zhang W, Sun X, Yoshimoto M, Chen Z, Zhu W, et al. Fkbp1a controls ventricular myocardium trabeculation and compaction by regulating endocardial Notch1 activity. Development. 2013;140(9):1946-1957
  92. 92. Hirono K, Saito K, Munkhsaikhan U, Xu F, Wang C, Lu L, et al. Familial left ventricular non-compaction is associated with a rare p.V407I variant in bone morphogenetic protein 10. Circulation Journal. 2019;83(8):1737-1746
  93. 93. Vatta M, Mohapatra B, Jimenez S, Sanchez X, Faulkner G, Perles Z, et al. Mutations in Cypher/ZASP in patients with dilated cardiomyopathy and left ventricular non-compaction. Journal of the American College of Cardiology. 2003;42(11):2014-2027
  94. 94. Schultze-Berndt A, Kuhnisch J, Herbst C, Seidel F, Al-Wakeel-Marquard N, Dartsch J, et al. Reduced systolic function and not genetic variants determine outcome in pediatric and adult left ventricular noncompaction cardiomyopathy. Frontiers in Pediatrics. 2021;9:722926
  95. 95. Armel TZ, Leinwand LA. Mutations in the beta-myosin rod cause myosin storage myopathy via multiple mechanisms. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(15):6291-6296
  96. 96. Arbustini E, Favalli V, Narula N, Serio A, Grasso M. Left ventricular noncompaction: A distinct genetic cardiomyopathy? Journal of the American College of Cardiology. 2016;68(9):949-966
  97. 97. Monserrat L, Hermida-Prieto M, Fernandez X, Rodriguez I, Dumont C, Cazon L, et al. Mutation in the alpha-cardiac actin gene associated with apical hypertrophic cardiomyopathy, left ventricular non-compaction, and septal defects. European Heart Journal. 2007;28(16):1953-1961
  98. 98. Parent JJ, Towbin JA, Jefferies JL. Left ventricular noncompaction in a family with lamin A/C gene mutation. Texas Heart Institute Journal. 2015;42(1):73-76
  99. 99. Mogensen J, Kruse TA, Borglum AD. Assignment of the human cardiac troponin I gene (TNNI3) to chromosome 19q13.4 by radiation hybrid mapping. Cytogenetics and Cell Genetics. 1997;79(3-4):272-273
  100. 100. Muntoni F, Bonne G, Goldfarb LG, Mercuri E, Piercy RJ, Burke M, et al. Disease severity in dominant Emery Dreifuss is increased by mutations in both emerin and desmin proteins. Brain. 2006;129(Pt 5):1260-1268
  101. 101. Dautant A, Meier T, Hahn A, Tribouillard-Tanvier D, di Rago JP, Kucharczyk R. ATP synthase diseases of mitochondrial genetic origin. Frontiers in Physiology. 2018;9:329
  102. 102. Finsterer J, Stollberger C, Steger C, Cozzarini W. Complete heart block associated with noncompaction, nail-patella syndrome, and mitochondrial myopathy. Journal of Electrocardiology. 2007;40(4):352-354
  103. 103. Dorner A, Giessen S, Gaub R, Grosse Siestrup H, Schwimmbeck PL, Hetzer R, et al. An isoform shift in the cardiac adenine nucleotide translocase expression alters the kinetic properties of the carrier in dilated cardiomyopathy. European Journal of Heart Failure. 2006;8(1):81-89
  104. 104. Arndt AK, Schafer S, Drenckhahn JD, Sabeh MK, Plovie ER, Caliebe A, et al. Fine mapping of the 1p36 deletion syndrome identifies mutation of PRDM16 as a cause of cardiomyopathy. American Journal of Human Genetics. 2013;93(1):67-77
  105. 105. Bolling MC, Pas HH, de Visser M, Aronica E, Pfendner EG, van den Berg MP, et al. PLEC1 mutations underlie adult-onset dilated cardiomyopathy in epidermolysis bullosa simplex with muscular dystrophy. The Journal of Investigative Dermatology. 2010;130(4):1178-1181
  106. 106. Girolami F, Iascone M, Tomberli B, Bardi S, Benelli M, Marseglia G, et al. Novel alpha-actinin 2 variant associated with familial hypertrophic cardiomyopathy and juvenile atrial arrhythmias: A massively parallel sequencing study. Circulation. Cardiovascular Genetics. 2014;7(6):741-750
  107. 107. Lang T, Yu L, Tu Q , Jiang J, Chen Z, Xin Y, et al. Molecular cloning, genomic organization, and mapping of PRKAG2, a heart abundant gamma2 subunit of 5'-AMP-activated protein kinase, to human chromosome 7q36. Genomics. 2000;70(2):258-263
  108. 108. Schmitt JP, Kamisago M, Asahi M, Li GH, Ahmad F, Mende U, et al. Dilated cardiomyopathy and heart failure caused by a mutation in phospholamban. Science. 2003;299(5611):1410-1413
  109. 109. Haghighi K, Kolokathis F, Pater L, Lynch RA, Asahi M, Gramolini AO, et al. Human phospholamban null results in lethal dilated cardiomyopathy revealing a critical difference between mouse and human. The Journal of Clinical Investigation. 2003;111(6):869-876
  110. 110. Dellefave LM, Pytel P, Mewborn S, Mora B, Guris DL, Fedson S, et al. Sarcomere mutations in cardiomyopathy with left ventricular hypertrabeculation. Circulation. Cardiovascular Genetics. 2009;2(5):442-449
  111. 111. Fiorillo C, Astrea G, Savarese M, Cassandrini D, Brisca G, Trucco F, et al. MYH7-related myopathies: Clinical, histopathological and imaging findings in a cohort of Italian patients. Orphanet Journal of Rare Diseases. 2016;11(1):91
  112. 112. Hasegawa H, Komuro I. Mutations in sarcomere protein genes as a cause of heart failure. Nihon Rinsho. 2007;65(Suppl 4):267-272
  113. 113. D'Amato G, Luxan G, de la Pompa JL. Notch signalling in ventricular chamber development and cardiomyopathy. The FEBS Journal. 2016;283(23):4223-4237
  114. 114. Finsterer J, Stollberger C, Fazio G. Neuromuscular disorders in left ventricular hypertrabeculation/noncompaction. Current Pharmaceutical Design. 2010;16(26):2895-2904
  115. 115. Tang S, Batra A, Zhang Y, Ebenroth ES, Huang T. Left ventricular noncompaction is associated with mutations in the mitochondrial genome. Mitochondrion. 2010;10(4):350-357
  116. 116. Liu S, Bai Y, Huang J, Zhao H, Zhang X, Hu S, et al. Do mitochondria contribute to left ventricular non-compaction cardiomyopathy? New findings from myocardium of patients with left ventricular non-compaction cardiomyopathy. Molecular Genetics and Metabolism. 2013;109(1):100-106
  117. 117. Bleyl SB, Mumford BR, Brown-Harrison MC, Pagotto LT, Carey JC, Pysher TJ, et al. Xq28-linked noncompaction of the left ventricular myocardium: Prenatal diagnosis and pathologic analysis of affected individuals. American Journal of Medical Genetics. 1997;72(3):257-265
  118. 118. Saric A, Andreau K, Armand AS, Moller IM, Petit PX. Barth syndrome: From mitochondrial dysfunctions associated with aberrant production of reactive oxygen species to pluripotent stem cell studies. Frontiers in Genetics. 2015;6:359
  119. 119. Karkucinska-Wieckowska A, Trubicka J, Werner B, Kokoszynska K, Pajdowska M, Pronicki M, et al. Left ventricular noncompaction (LVNC) and low mitochondrial membrane potential are specific for Barth syndrome. Journal of Inherited Metabolic Disease. 2013;36(6):929-937
  120. 120. Hsieh YT, Tu HF, Yang MH, Chen YF, Lan XY, Huang CL, et al. Mitochondrial genome and its regulator TFAM modulates head and neck tumourigenesis through intracellular metabolic reprogramming and activation of oncogenic effectors. Cell Death & Disease. 2021;12(11):961
  121. 121. Westermann B. Bioenergetic role of mitochondrial fusion and fission. Biochimica et Biophysica Acta. 2012;1817(10):1833-1838
  122. 122. Otera H, Mihara K. Molecular mechanisms and physiologic functions of mitochondrial dynamics. Journal of Biochemistry. 2011;149(3):241-251
  123. 123. Dorn GW 2nd. Mitochondrial dynamism and heart disease: Changing shape and shaping change. EMBO Molecular Medicine. 2015;7(7):865-877
  124. 124. Rotig A, de Lonlay P, Chretien D, Foury F, Koenig M, Sidi D, et al. Aconitase and mitochondrial iron-sulphur protein deficiency in Friedreich ataxia. Nature Genetics. 1997;17(2):215-217
  125. 125. Kokoszka JE, Waymire KG, Levy SE, Sligh JE, Cai J, Jones DP, et al. The ADP/ATP translocator is not essential for the mitochondrial permeability transition pore. Nature. 2004;427(6973):461-465
  126. 126. Chen H, Detmer SA, Ewald AJ, Griffin EE, Fraser SE, Chan DC. Mitofusins Mfn1 and Mfn2 coordinately regulate mitochondrial fusion and are essential for embryonic development. The Journal of Cell Biology. 2003;160(2):189-200
  127. 127. Finsterer J. Genetic, pathogenetic, and phenotypic implications of the mitochondrial A3243G tRNALeu(UUR) mutation. Acta Neurologica Scandinavica. 2007;116(1):1-14
  128. 128. Shanske S, Pancrudo J, Kaufmann P, Engelstad K, Jhung S, Lu J, et al. Varying loads of the mitochondrial DNA A3243G mutation in different tissues: Implications for diagnosis. American Journal of Medical Genetics. Part A. 2004;130A(2):134-137
  129. 129. Finsterer J, Stollberger C, Wegmann R, Janssen LA. Acquired left ventricular hypertrabeculation/noncompaction in myotonic dystrophy type 1. International Journal of Cardiology. 2009;137(3):310-313
  130. 130. de Andrade PB, Rubi B, Frigerio F, van den Ouweland JM, Maassen JA, Maechler P. Diabetes-associated mitochondrial DNA mutation A3243G impairs cellular metabolic pathways necessary for beta cell function. Diabetologia. 2006;49(8):1816-1826
  131. 131. Mkaouar-Rebai E, Kammoun F, Chamkha I, Kammoun N, Hsairi I, Triki C, et al. A de novo mutation in the adenosine triphosphatase (ATPase) 8 gene in a patient with mitochondrial disorder. Journal of Child Neurology. 2010;25(6):770-775
  132. 132. Jonckheere AI, Hogeveen M, Nijtmans LG, van den Brand MA, Janssen AJ, Diepstra JH, et al. A novel mitochondrial ATP8 gene mutation in a patient with apical hypertrophic cardiomyopathy and neuropathy. Journal of Medical Genetics. 2008;45(3):129-133

Written By

Gowthami Mahendran and Margaret A. Schwarz

Submitted: 24 June 2022 Reviewed: 22 November 2022 Published: 20 December 2022