Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2733 2023-04-07 00:57:04 |
2 format change Meta information modification 2733 2023-04-07 03:51:30 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Lau, C.; Gul, U.; Liu, B.; Captur, G.; Hothi, S.S. Cardiovascular Magnetic Resonance Imaging in Familial Dilated Cardiomyopathy. Encyclopedia. Available online: https://encyclopedia.pub/entry/42857 (accessed on 01 July 2024).
Lau C, Gul U, Liu B, Captur G, Hothi SS. Cardiovascular Magnetic Resonance Imaging in Familial Dilated Cardiomyopathy. Encyclopedia. Available at: https://encyclopedia.pub/entry/42857. Accessed July 01, 2024.
Lau, Clement, Uzma Gul, Boyang Liu, Gabriella Captur, Sandeep S. Hothi. "Cardiovascular Magnetic Resonance Imaging in Familial Dilated Cardiomyopathy" Encyclopedia, https://encyclopedia.pub/entry/42857 (accessed July 01, 2024).
Lau, C., Gul, U., Liu, B., Captur, G., & Hothi, S.S. (2023, April 07). Cardiovascular Magnetic Resonance Imaging in Familial Dilated Cardiomyopathy. In Encyclopedia. https://encyclopedia.pub/entry/42857
Lau, Clement, et al. "Cardiovascular Magnetic Resonance Imaging in Familial Dilated Cardiomyopathy." Encyclopedia. Web. 07 April, 2023.
Cardiovascular Magnetic Resonance Imaging in Familial Dilated Cardiomyopathy
Edit

Dilated cardiomyopathy (DCM) is a common cause of non-ischaemic heart failure, conferring high morbidity and mortality, including sudden cardiac death due to systolic dysfunction or arrhythmic sudden death. Within the DCM cohort exists a group of patients with familial disease.

familial dilated cardiomyopathy cardiovascular magnetic resonance imaging DCM CMR

1. Introduction

Cardiomyopathies are myocardial disorders in which the heart is structurally and functionally abnormal. They are currently sub-classified on the basis of cardiac morphology as hypertrophic cardiomyopathy (HCM), dilated cardiomyopathy (DCM), arrhythmogenic cardiomyopathy (ACM), and left ventricular noncompaction cardiomyopathy (LVNC). The European Society of Cardiology (ESC) divides dilated cardiomyopathy into two groups, familial and nonfamilial [1]. Conventionally, idiopathic DCM with at least one first- or second-degree relative with confirmed idiopathic DCM is defined as familial DCM [1][2]. These non-ischaemic cardiomyopathies are less common than ischaemic cardiomyopathy [3]. Of non-ischaemic aetiologies, idiopathic DCM is the second most common aetiology, accounting for 31% of cases. Non-ischaemic DCM is more common in female, non-white, and younger individuals [3].
Dilated cardiomyopathy accounts for up to one-third of heart failure cases and is one of the leading causes for cardiac transplantation. The reported prevalence of DCM from epidemiological data is 36.5/100,000 individuals in Western populations [3]. This is likely to be an underestimate, however, since its diagnosis has relied on modalities with low sensitivity, such as echocardiography or angiography. Hershberger and colleagues, in their review, were able to report a higher prevalence of DCM of > 1 per 250 individuals on the basis of recent data [1][4].
Estimations of the prevalence of familial DCM range widely, between 2% to 65%, averaging 23% in a meta-analysis of 23 studies [5]. This is due partly to heterogeneity of the diagnostic criteria as well as increasing diagnosis over time related to more systematic clinical screening [5]. Familial DCM has the following subtypes with already mapped genetic loci (>40): autosomal dominant, autosomal recessive, X-linked, and mitochondrial forms. These might comprise either a pure cardiomyopathy or may have associated myopathy [6][7]. The penetration of familial DCMs is incomplete, variable, and age-dependent [6][7].
Amongst familial DCM, monogenic causes account only for approximately 30–40% of cases [8][9]. The implication of this is that that traditional Mendelian considerations will leave more than half of cases without a defined monogenic cause. It is, therefore, likely that complex mechanisms underlie familiar DCM rather than monogenic transmission alone. It has accordingly been proposed that common variants predispose to DCM in the appropriate environmental exposure, while rarer variants may underlie monogenic forms. Including rare variants in the genetic panel increases the yield of genetic testing for DCM, with a genetic diagnosis achieved in approximately 40% of apparently familial cases [4]. Pathogenic genetic variants can be identified in 15–25% of sporadic DCM [8][9]. Titin (TTN) mutations are the most common aetiology of familial DCM, occurring in ~25% of familial cases of DCM and in 18% of sporadic cases [1].
Rare variants in more than 30 genes can produce a DCM phenotype, some of which also underlie other cardiomyopathies, inherited muscle diseases, or myopathic syndromes. These genes encode both contractile and non-contractile proteins, such as cytoskeletal proteins, abnormalities of which result in the DCM phenotype. This results in reduced resistance to mechanical stress as well as abnormalities of intracellular calcium handling, myocellular energetics, and sarcolemmal ion channel function [1]. De novo mutations are rare and defined when none of the biological parents carry the offspring’s mutation and confirm the pathogenic status of genetic variants [9]. A multicentre study tested the hypothesis that both familial and non-familial DCM have a rare variant genetic basis and concluded that most idiopathic DCM have a genetic basis [10].

2. Diagnosis

The diagnostic process consists of a detailed clinical history alongside at least a three-generation family pedigree, comprehensive cardiac imaging, biochemical profile, and genetic testing, where clinically indicated. Systematic screening with electrocardiography (ECG) and echocardiography of first-degree relatives of patients with idiopathic DCM has been proposed to identify subclinical forms. European Society of Cardiology Guidelines recommend screening with an ECG and echocardiogram in all first-degree relatives of an index patient with DCM, irrespective of family history [11].
Mestroni et al. [6] define familial DCM by either the presence of two or more affected relatives within a single family or in the presence of a first-degree relatives of a DCM patient with SCD below age 35 years [6]. Criteria proposed to diagnose the index case includes imaging criteria, family history, ECG, imaging criteria, and exclusion of competing causes, such as significant coronary artery disease, chronic alcohol excess, uncontrolled hypertension, persistent arrhythmia, pericardial disease, congenital heart disease, and cor pulmonale. There are some inherent difficulties in identifying the index case. Firstly, the varying clinical presentation and course of the disease lends diagnostic challenges. Secondly, acquired disorders, such as hypertension, excess alcohol intake, and systemic inflammatory diseases may produce phenocopies of idiopathic DCM or act as environmental factors, unmasking rare variants [6].

3. Genetics in Familial DCMs

Studying the genetic basis of DCM requires either multigeneration DCM pedigrees or genome-wide sequencing. Most studies use the latter approach, and once DCM-associated variants are identified, the numbers of DCM patients with these variants are compared with the number of individuals in the reference datasets carrying the same variants. A probability of 90% or more is required to be labelled pathogenetic, although this does not necessarily reflect causation [1]. Sequencing of large numbers of genes is required due to low prevalence, heterogeneity of mutations, private mutations, modifier genes, and different mutations producing the same phenotype [4]. Fortunately, there are large, easily available genetic datasets which enable evaluation of already identified pathogenic mutations. Another approach, used by most clinical centres, is to perform targeted next generation sequencing of high-risk variants.
Several genome-wide sequencing association studies (GWAS) have identified genetic variants associated with DCM. However, the yield has been limited by modest sample sizes (< 5000 cases). The prevalence of these pathogenic genetic variants is greater than the estimated disease prevalence. Hershberger suggested this mismatch is due either to lower penetrance of the mutations, non-pathogenicity of some, or that the actual DCM prevalence is higher than estimated [4]. Villard et al. [12] were the first to perform genome-wide study, while Esslinger et al. [13] and Meder et al. [14] carried out similar studies involving 3000 and 4000 DCM cases with matched controls, respectively.

4. Cardiac Imaging in Familial DCMs

DCM has been defined by echocardiography by the degree of systolic impairment, that is, fractional shortening (FS) less than 25% (> 2SD) and/or ejection fraction less than 45% (> 2SD) and the degree of LV enlargement; or LV end diastolic diameter (LVEDD) greater than 117% (2SD (112%) plus 5%) or end diastolic volume (LVEDV) greater than 2SD of the predicted value, as corrected for age and body surface area, excluding any known cause resulting in the myocardial abnormality observed [6].
Echocardiography is often the first imaging test for assessing LV remodelling and also provides associated data, such as the presence and severity of functional mitral regurgitation. Speckle-tracking echocardiography uses the distinct speckle pattern in the myocardium to assess myocardial deformation. Abnormalities of strain and strain rate can be detected by echocardiography in first-degree relatives of patients with DCM, indicating a subclinical phenotype [15].
Cardiovascular magnetic resonance imaging (CMR) is the reference standard for measurement of ventricular volume, ejection fraction, and myocardial mass. In addition, CMR detects myocardial oedema, which, when present, may suggest an inflammatory basis for the observed phenotype. Long native myocardial T1 time and high extracellular volume (ECV) fraction may be helpful in differentiating DCM from athletic heart adaptation or iron overload cardiomyopathy. The presence, pattern, and burden of late gadolinium enhancement (LGE) may be helpful in determining the risk of malignant ventricular arrhythmias. Echocardiography may be suboptimal in certain individuals when CMR is recommended as an alternative. Progressive increases in chamber dimensions, strain abnormalities, and LGE are features of early DCM [16]. Longitudinal studies over many years with imaging are required to characterise DCM progression in genetically predisposed individuals. CMR offers higher repeatability in volumes and ejection fraction and may allow detection of subtle changes in surveillance of mutation carriers. LGE detects replacement fibrosis but not diffuse fibrosis, so it may be deceptively reassuring even in those with established diffuse fibrosis, where native T1 is long and ECV is high.
Amin et al. [2] demonstrated that the combining CMR with genetic information allows better DCM stratification and results in a change in management, as per an ESC Position Paper [2]. In DCM, the degree of fibrosis shown by LGE, is a predictor of mortality and hospitalisation, particularly ventricular arrhythmias [17]. Hyper-trabeculation is also detected commonly in DCM (36%), although its presence was not associated with adverse outcomes, and this can be detected in normal hearts [18][19].
Imaging traits of DCM have been used to investigate genetic variants involved in a DCM phenotype [18][20]. valuation of approximately six thousand subjects from cardiac imaging from the Candidate Gene Association Resource (CARe) Study and more than a thousand individuals from the Multi-Ethnic Study of Atherosclerosis (MESA) with CMR yielded four associated genes [21]. Pirrucello et al. [7] analysed CMR-derived left ventricular measurements in 36,000 UK Biobank and 2000 Multi-Ethnic Study of Atherosclerosis participants, identified 45 new loci, and developed a polygenic score for prediction of DCM on the basis of variants most strongly associated with DCM phenotypic variables on CMR.

5. CMR Sequences for DCM Assessment

The cardiac MRI protocol for DCM involves cine imaging and tissue characterisation, including mapping and gadolinium-enhanced images; flow mapping, oedema, and stress perfusion imaging may be considered. Cine images use a balanced steady-state free processing (SSFP) sequence, which provides high signal-to-noise ratio and myocardial blood pool contrast or flash (spoiled gradient echo) sequence in case of cardiac device or at 3T. These are usually retrospectively ECG gated along with breath-held-in expiration, with a minimum of 25 reconstructed phases, with a slice thickness and gap of 8 mm and 2 mm to give temporal resolution less than or equal to 45 ms and spatial resolution 2 mm.
Gadolinium-enhanced late images are inversion recovery or phase-sensitive inversion recovery gradient echo sequences, segmented as a routine, while single shot for patients with difficulty holding their breath or irregular rhythm. In plane resolution, it should be ~1.4–1.8 mm. These are generally acquired 10 min after the contrast injection; however, the delay should be tailored to the dose and protocol; images are acquired every other beat but can vary according to heart rate. A TI scout can be used as a guide for choosing inversion time to null myocardium; however, inversion time can be different due to different readout parameters. The late gadolinium-enhanced images are acquired in the same planes and slices as cine images.
T1 mapping sequences most frequently involves shortened MOLLI (ShMOLLI) during breath-holding, with the trigger delay set to end-diastole, slice thickness 6–8 mm, with in-plane resolution 1.6–2 mm. The native and 15 min post contrast maps are acquired as short axis slice, but number and plane of slices can be modified. Other sequences, such as SASHA, which involve saturation instead of inversion, are also available.
T2 mapping uses either a T2-prepared SSFP sequence or fast spin echo sequences, usually with motion correction.
Black blood T2-weighted short tau inversion recovery (STIR) sequence is used for oedema imaging. Images are usually acquired in three short axis slices as well as long axis views.

6. CMR Characteristics of Familial DCMs

Septal linear mid-wall fibrosis is characteristic of, although not specific to, familial DCM. Furthermore, its presence carries adverse prognosis [22]. In a meta-analysis by Becker et al., LGE was present in approximately 45% of patients with DCM and was associated with adverse outcomes, including ventricular arrhythmia, hospitalisation, and death, while its absence predicted LV reverse remodelling [20]. Table 1 summarises the features of genetic DCM subtypes, which are now detailed further by subtype.
Table 1. Genetic dilated cardiomyopathies and their clinical, MRI, and arrhythmia features.

Mutation

Screening and Transmission

Age of Presentation/Prognosis

ECG and Clinical Features

Extra-Cardiac Features

Imaging Features/Volume and Function

Tissue Characterization

Arrhythmia/Conduction Disease Risk

Lamin

Recommended commencing 10–12 years by ECG and Echo

Autosomal dominant

Young age onset and progression

Conduction or depolarization abnormalities, such as IVCD/BBB/

Early abnormalities in strain or diastolic function

Arterial and venous thromboembolism

Isolated LV dilation or dysfunction; marked dilation unusual

Wall thinning not present

RWMAs (basal) in Lamin A/C

HNDCM to DCM progress

Prominent right ventricular epicardial fat

Early increase in ECV

LGE shows basal to mid septal, mid-myocardial fibrosis (extensive fibrosis only late in disease)

Increased arrhythmias and both AV and SA nodal conduction disease

Dystrophin

(DMD and BMD)

Cardiac screening recommended in mutation carrier females along with affected males

Consider CMR in screening alongside ECG and echo

X linked

Childhood onset, progression from HNDCM to typical DCM.

BMD has an earlier and more severe disease course than DMD

Increased right precordial ECG R-to-S ratio

Deep Q waves in the lateral leads

Early abnormalities in strain

Typical skeletal muscle weakness and disability in affected

May be none in carriers

Wall thinning and RWMAs lateral wall, inferior, or septum

Early increased ECV, while still normal function

DCM or HCM phenotype

Mid-wall or subepicardial fibrosis in hypokinetic segments (inferolateral) in early disease (sometimes with preserved LV function)

Fibrosis becomes extensive or transmural in advanced stages

Conduction abnormalities and tachyarrhythmias (both SVT and VT)

Early onset in severe forms

Dystrophin

(EDMD and others)

Screening recommended

 

Early strain abnormalities

Typical skeletal muscle weakness and disability

DCM phenotype but no specific features

Fibrosis not early feature in EDMD

Septal mid-wall fibrosis early feature in limb–girdle type

Tachyarrhythmias and conduction abnormalities

Sodium channel

           

Arrhythmias

Filamin C

Cardiac screening recommended in mutation-positive or first-degree relatives of proband

Consider CMR

Autosomal

 

Low voltage and flat or inverted T waves inferolaterally

Early LGE with normal echo

ECG features of ACM absent in ACM phenotype

 

DCM or ACM (LV) phenotype

Subepicardial or mid-myocardial inferolateral fibrosis

“Ring-like” circumferential fibrosis, but not in all cases

Ventricular arrhythmias with a more marked and malignant course than average DCM cases

Sarcomere

Screening recommended early age

Exact gene determines inheritance mode and penetration

Severe and progressive disease if childhood onset

Mild and non-progressive if adult onset

Indistinguishable from others since no red flag features

 

DCM or HCM phenotype

Mid-wall, linear fibrosis may or may not be present.

 

Desmin

Screening recommended

Autosomal dominant

CMR may be considered alongside ECG and Echo

Variable onset and prognosis depending on exact mutation

 

May or may not have skeletal muscle involvement

RCM, ACM, or DCM phenotype

Focal wall hypertrophy early disease

Oedema in acute phase

Perfusion defect

Focal fibrosis by LGE (mid-wall or subepicardial) anywhere in LV (apex, anterior, septal, lateral, or inferior) or RV

Transmural scar in advanced stage disease

AV conduction abnormalities and ventricular arrhythmias

Danon

X-linked dominant

   

Syndromic facial features

HCM or DCM phenotype

LGE scar can be extensive and spares the mid-septum.

High ECV/T1

 

Titin

Cardiac screening recommended affected and carriers

Autosomal Dominant

   

Skeletal muscle, syndrome features

CHD or DCM

Thinner walls, lower LVEF

 

Arrhythmias

Mitochondrial

Autosomal recessive, maternally inherited or dominant

   

Neurological, endocrine, and ophthalmic

HCM or DCM phenotype depending on type of disease

Diffuse fibrosis (T1/ECV)

Extensive fibrosis only I end stage

Conduction abnormalities and Wolf Parkinson White syndrome

AVB prevalent in Kearns Sayre

7. Role of CMR in Guiding Device Therapy

In patients with DCM, the presence of LGE in CMR coupled with an LVEF ≤ 50% suggests consideration of an implantable cardiac defibrillator (ICD) for primary prevention [23]. In a study of 452 patients with non-ischaemic cardiomyopathy and LVEF < 35% on optimal medical therapy who met the criteria for ICD insertion, ICD reduced all-cause mortality (HR, 0.45; 95% CI: 0.26–0.77) and cardiovascular death (HR, 0.51; 95% CI: 0.27–0.97) when LGE was present after a median follow-up period of 37.9 months [24]. The DANISH-MRI study, a pre-specified sub-study of the DANISH trial (152), recruited 252 patients with non-ischemic DCM and an indication for primary-prevention ICD on optimal medical management [25]. Overall prognosis in the patients with LV scarring was worse, but ICD implantation did not significantly reduce all-cause mortality in those with LV scarring compared with those without LV scarring (HR, 1.18; 95% CI: 0.59–2.38 vs. HR 1.00; 95% CI: 0.39–2.53, p for interaction = 0.79) [25].
CMR can also be used to predict and guide response to device therapy. Chalil et al. [26] derived the tissue synchronization index (CMR-TSI) determined by segmental radial wall motion data, generating tissue synchronization polar maps of the LV, which is a global dyssynchrony measure. A CMR-TSI ≥ 110 ms was an independent predictor of death or unplanned hospitalisation in a cohort of 77 HF patients referred for cardiac resynchronisation therapy (CRT) after a median follow-up of 2 years [26]. The presence of LGE may also guide lead placement in CRT. In 60 patients receiving CRT implantation, RV and LV leads over scarring was associated with non-response to CRT [27]. LV lead positions at areas of LGE compared with viable myocardium were associated with an increased risk of CV death (HR 6.34; p < 0.0001) or HF hospitalizations (HR 5.57; p < 0.0001) [28].

References

  1. Schultheiss, H.P.; Fairweather, D.L.; Caforio, A.L.P.; Escher, F.; Hershberger, R.E.; Lipshultz, S.E.; Liu, P.P.; Matsumori, A.; Mazzanti, A.; McMurray, J.; et al. Dilated Cardiomyopathy. Nat. Rev. Dis. Prim. 2019, 5, 32.
  2. Amin, R.; Jones, R.; Hammersley, D.; Gati, S.; Khalique, Z.; Almogheer, B.; Pennell, D.J.; Pantazis, A.; Baksi, J.; Prasad, S.K.; et al. Abstract 15590: Genetic Testing and Cardiac Magnetic Resonance Imaging Results Improve Precision Stratification in Dilated Cardiomyopathy. Circulation 2020, 142, A15590.
  3. McNally, E.M.; Mestroni, L. Dilated Cardiomyopathy. Circ. Res. 2017, 121, 731–748.
  4. Hershberger, R.E.; Hedges, D.J.; Morales, A. Dilated Cardiomyopathy: The Complexity of a Diverse Genetic Architecture. Nat. Rev. Cardiol. 2013, 10, 531–547.
  5. Petretta, M.; Pirozzi, F.; Sasso, L.; Paglia, A.; Bonaduce, D. Review and Metaanalysis of the Frequency of Familial Dilated Cardiomyopathy. Am. J. Cardiol. 2011, 108, 1171–1176.
  6. Mestroni, L.; Maisch, B.; McKenna, W.J.; Schwartz, K.; Charron, P.; Rocco, C.; Tesson, F.; Richter, A.; Wilke, A.; Komajda, M. Guidelines for the Study of Familial Dilated Cardiomyopathies. Collaborative Research Group of the European Human and Capital Mobility Project on Familial Dilated Cardiomyopathy. Eur. Heart J. 1999, 20, 93–102.
  7. Pirruccello, J.P.; Bick, A.; Wang, M.; Chaffin, M.; Friedman, S.; Yao, J.; Guo, X.; Venkatesh, B.A.; Taylor, K.D.; Post, W.S.; et al. Analysis of Cardiac Magnetic Resonance Imaging in 36,000 Individuals Yields Genetic Insights into Dilated Cardiomyopathy. Nat. Commun. 2020, 11, 2254.
  8. Ganesh, S.K.; Arnett, D.K.; Assimes, T.L.; Basson, C.T.; Chakravarti, A.; Ellinor, P.T.; Engler, M.B.; Goldmuntz, E.; Herrington, D.M.; Hershberger, R.E.; et al. Genetics and Genomics for the Prevention and Treatment of Cardiovascular Disease: Update. Circulation 2013, 128, 2813–2851.
  9. Rosenbaum, A.N.; Agre, K.E.; Pereira, N.L. Genetics of Dilated Cardiomyopathy: Practical Implications for Heart Failure Management. Nat. Rev. Cardiol. 2020, 17, 286–297.
  10. Kinnamon, D.D.; Morales, A.; Bowen, D.J.; Burke, W.; Hershberger, R.E.; Morales, A.; Bowen, D.J.; Gastier-Foster, J.M.; Nickerson, D.A.; Dorschner, M.O.; et al. Toward Genetics-Driven Early Intervention in Dilated Cardiomyopathy: Design and Implementation of the DCM Precision Medicine Study. Circ. Cardiovasc. Genet. 2017, 10, e001826.
  11. Wilde, A.A.M.; Semsarian, C.; Márquez, M.F.; Shamloo, A.S.; Ackerman, M.J.; Ashley, E.A.; Sternick, E.B.; Barajas-Martinez, H.; Behr, E.R.; Bezzina, C.R.; et al. European Heart Rhythm Association (EHRA)/Heart Rhythm Society (HRS)/Asia Heart Rhythm Society (APHRS)/Latin American Heart Rhythm Society (LAHRS) Expert Statement on the State of Genetic Testing for Cardiac Diseases. Europace 2022, 24, 1307.
  12. Villard, E.; Perret, C.; Gary, F.; Proust, C.; Dilanian, G.; Hengstenberg, C.; Ruppert, V.; Arbustini, E.; Wichter, T.; Germain, M.; et al. A Genome-Wide Association Study Identifies Two Loci Associated with Heart Failure Due to Dilated Cardiomyopathy. Eur. Heart J. 2011, 32, 1065–1076.
  13. Esslinger, U.; Garnier, S.; Korniat, A.; Proust, C.; Kararigas, G.; Müller-Nurasyid, M.; Empana, J.P.; Morley, M.P.; Perret, C.; Stark, K.; et al. Exome-Wide Association Study Reveals Novel Susceptibility Genes to Sporadic Dilated Cardiomyopathy. PLoS ONE 2017, 12, e0172995.
  14. Meder, B.; Rühle, F.; Weis, T.; Homuth, G.; Keller, A.; Franke, J.; Peil, B.; Bermejo, J.L.; Frese, K.; Huge, A.; et al. A Genome-Wide Association Study Identifies 6p21 as Novel Risk Locus for Dilated Cardiomyopathy. Eur. Heart J. 2014, 35, 1069–1077.
  15. Cordero-Reyes, A.M.; Youker, K.; Estep, J.D.; Torre-Amione, G.; Nagueh, S.F. Molecular and Cellular Correlates of Cardiac Function in End-Stage DCM: A Study Using Speckle Tracking Echocardiography. JACC Cardiovasc. Imaging 2014, 7, 441–452.
  16. Japp, A.G.; Gulati, A.; Cook, S.A.; Cowie, M.R.; Prasad, S.K. The Diagnosis and Evaluation of Dilated Cardiomyopathy. J. Am. Coll. Cardiol. 2016, 67, 2996–3010.
  17. Gao, C.; Tao, Y.; Pan, J.; Shen, C.; Zhang, J.; Xia, Z.; Wan, Q.; Wu, H.; Gao, Y.; Shen, H.; et al. Evaluation of Elevated Left Ventricular End Diastolic Pressure in Patients with Preserved Ejection Fraction Using Cardiac Magnetic Resonance. Eur. Radiol. 2019, 29, 2360–2368.
  18. Amzulescu, M.S.; Rousseau, M.F.; Ahn, S.A.; Boileau, L.; de Meester De Ravenstein, C.; Vancraeynest, D.; Pasquet, A.; Vanoverschelde, J.L.; Pouleur, A.C.; Gerber, B.L. Prognostic Impact of Hypertrabeculation and Noncompaction Phenotype in Dilated Cardiomyopathy: A CMR Study. JACC Cardiovasc. Imaging 2015, 8, 934–946.
  19. Dawson, D.K.; McLernon, D.J.; Raj, V.J.; Maceira, A.M.; Prasad, S.; Frenneaux, M.P.; Pennell, D.J.; Kilner, P.J. Cardiovascular Magnetic Resonance Determinants of Left Ventricular Noncompaction. Am. J. Cardiol. 2014, 114, 456–462.
  20. Becker, M.A.J.; Cornel, J.H.; van de Ven, P.M.; van Rossum, A.C.; Allaart, C.P.; Germans, T. The Prognostic Value of Late Gadolinium-Enhanced Cardiac Magnetic Resonance Imaging in Nonischemic Dilated Cardiomyopathy: A Review and Meta-Analysis. JACC Cardiovasc. Imaging 2018, 11, 1274–1284.
  21. Fox, E.R.; Musani, S.K.; Barbalic, M.; Lin, H.; Yu, B.; Ogunyankin, K.O.; Smith, N.L.; Kutlar, A.; Glazer, N.L.; Post, W.S.; et al. Genome-Wide Association Study of Cardiac Structure and Systolic Function in African Americans: The Candidate Gene Association Resource (CARe) Study. Circ. Cardiovasc. Genet. 2013, 6, 37–46.
  22. Van Berlo, J.H.; de Voogt, W.G.; van der Kooi, A.J.; van Tintelen, J.P.; Bonne, G.; Yaou, R.B.; Duboc, D.; Rossenbacker, T.; Heidbüchel, H.; de Visser, M.; et al. Meta-Analysis of Clinical Characteristics of 299 Carriers of LMNA Gene Mutations: Do Lamin A/C Mutations Portend a High Risk of Sudden Death? J. Mol. Med. 2004, 83, 79–83.
  23. Zeppenfeld, K.; Tfelt-Hansen, J.; de Riva, M.; Winkel, B.G.; Behr, E.R.; Blom, N.A.; Charron, P.; Corrado, D.; Dagres, N.; de Chillou, C.; et al. 2022 ESC Guidelines for the Management of Patients with Ventricular Arrhythmias and the Prevention of Sudden Cardiac Death. Eur. Heart J. 2022, 43, 3997–4126.
  24. Gutman, S.J.; Costello, B.T.; Papapostolou, S.; Voskoboinik, A.; Iles, L.; Ja, J.; Hare, J.L.; Ellims, A.; Kistler, P.M.; Marwick, T.H.; et al. Reduction in Mortality from Implantable Cardioverter-Defibrillators in Non-Ischaemic Cardiomyopathy Patients Is Dependent on the Presence of Left Ventricular Scar. Eur. Heart J. 2019, 40, 542–550.
  25. Elming, M.B.; Hammer-Hansen, S.; Voges, I.; Nyktari, E.; Raja, A.A.; Svendsen, J.H.; Pehrson, S.; Signorovitch, J.; Køber, L.; Prasad, S.K.; et al. Myocardial Fibrosis and the Effect of Primary Prophylactic Defibrillator Implantation in Patients with Non-Ischemic Systolic Heart Failure-DANISH-MRI. Am. Heart J. 2020, 221, 165–176.
  26. Chalil, S.; Stegemann, B.; Muhyaldeen, S.; Khadjooi, K.; Smith, R.E.A.; Jordan, P.J.; Leyva, F. Intraventricular Dyssynchrony Predicts Mortality and Morbidity After Cardiac Resynchronization Therapy: A Study Using Cardiovascular Magnetic Resonance Tissue Synchronization Imaging. J. Am. Coll. Cardiol. 2007, 50, 243–252.
  27. Wong, T.C.; Piehler, K.M.; Zareba, K.M.; Lin, K.; Phrampus, A.; Patel, A.; Moon, J.C.; Ugander, M.; Valeti, U.; Holtz, J.E.; et al. Myocardial Damage Detected by Late Gadolinium Enhancement Cardiovascular Magnetic Resonance Is Associated with Subsequent Hospitalization for Heart Failure. J. Am. Heart Assoc. 2013, 2, e000416.
  28. Leyva, F.; Foley, P.W.X.; Chalil, S.; Ratib, K.; Smith, R.E.A.; Prinzen, F.; Auricchio, A. Cardiac Resynchronization Therapy Guided by Late Gadolinium-Enhancement Cardiovascular Magnetic Resonance. J. Cardiovasc. Magn. Reson. 2011, 13, 29.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , ,
View Times: 356
Revisions: 2 times (View History)
Update Date: 07 Apr 2023
1000/1000
Video Production Service