Elsevier

Heart Rhythm

Volume 19, Issue 3, March 2022, Pages 497-504
Heart Rhythm

Contemporary Review
An overview of heart rhythm disorders and management in myotonic dystrophy type 1

https://doi.org/10.1016/j.hrthm.2021.11.028Get rights and content

Myotonic dystrophy type 1 (DM1) is the most common adult form of muscular dystrophy, presenting with a constellation of systemic findings secondary to a CTG triplet expansion of the noncoding region of the DMPK gene. Cardiac involvement is frequent, with conduction disease and supraventricular and ventricular arrhythmias being the most prevalent cardiac manifestations, often developing from a young age. The development of cardiac arrhythmias has been linked to increased morbidity and mortality, with sudden cardiac death well described. Strategies to mitigate risk of arrhythmic death have been developed. In this review, we outline the current knowledge on the pathophysiology of rhythm abnormalities in patients with myotonic dystrophy and summarize available knowledge on arrhythmic risk stratification. We also review management strategies from an electrophysiological perspective, attempting to underline the substantial unmet need to address residual arrhythmic risks for this population.

Introduction

Myotonic dystrophy type 1 (DM1; Steinert disease) is the most common form of adult muscular dystrophy, with a prevalence of 1 in 8000 patients. It is characterized by clinical features of striated muscle weakness; early myotonia; ocular, cutaneous, pulmonary, central nervous system, and metabolic abnormalities; and cardiac manifestations (Supplemental Table 1).1 The genetic basis of DM1 is an unstable CTG triplet repeat expansion in the DMPK gene in the 3ʹ noncoding region located in chromosome 19q13.3. Repeats greater than 39 CTGs are always associated with clinical disease.2 Inheritance is autosomal dominant, and genetic anticipation occurs, as with other triplet repeat disorders, associated with worsening phenotypic severity with sequential generations.2 The onset of phenotypic expression varies significantly, from a more severe congenital form to mild, late-onset disease. Disease severity and age of onset correlate poorly with expansion size as measured in white blood cells, probably due to somatic mosaicism.3 Myotonic dystrophy type 2 (DM2), which stems from CCTG repeats in the nucleic acid-binding protein (CNBP) gene, is rarer than DM1, with a prevalence that currently is undefined. Both types share similarities in pathophysiology and phenotypical expression. Patients with DM2 develop cardiac abnormalities similar to those of their type 1 counterparts but at an older age. Cardiac management in DM2 patients is largely extrapolated from that of DM1 patients.4 Whether this practice is valid in its entirety remains unresolved due to the lack of adequate data. For the purpose of this review, we will focus on data derived from DM1 patients.

Cardiac involvement is a well-recognized driver of mortality in myotonic dystrophy, in some cohorts accounting for up to one-third of events.5 It is manifested mainly by conduction disease and the propensity to supraventricular and ventricular arrhythmias (Supplemental Table 2). Younger patients exhibit higher risk for potentially life-threatening cardiac disease than their general population peers, and the risk tends to increase with time due to the progressive nature of the condition.5, 6, 7, 8 A minority of patients may develop systolic dysfunction that is associated with increased risk for life-threatening arrhythmias.9 Considering that reduced mobility may conceal heart failure symptoms, supraventricular arrhythmias have the propensity for slower ventricular response rates, and advanced conduction disturbances may be intermittent, the presence of asymptomatic systolic dysfunction and subclinical conduction system involvement may remain undetected. In this setting, the role of cardiology, and particularly electrophysiology, in the management of this population becomes increasingly relevant.

Section snippets

Analogies of DM1 to ion channel disorders

Although cardiac disease is a well-recognized complication of DM1 in knockout mice and humans, the mechanism by which CTG expansions lead to cardiac manifestations is not entirely clear. Increasing size of CTG repeats is considered to affect splicing, either by reducing transcription or, more likely, resulting in abnormal DMPK isoforms, with consequent impaired localization in specific tissue cytoplasm.10

DMPK gene translation produces an 80-kD serine/threonine protein kinase that is highly

Conduction disease in DM1

The observation that patients with DM developed conduction abnormalities was made early in the 20th century, with researchers highlighting advanced-degree AV block as the principal feature in DM1-related cardiac involvement.33 Electrophysiological testing used early in the investigation of DM1 patients demonstrated a high prevalence of His-Purkinje abnormalities, as reflected by HV interval prolongation.34 The progressive nature of conduction defects was confirmed in small cohorts, utilizing

Supraventricular arrhythmias in DM1

Supraventricular tachyarrhythmias represent the second most frequent heart rhythm abnormality in DM1 patients and are considered to substantially contribute to cardiac mortality and morbidity.38 Rapidly conducted atrial fibrillation (AF) and flutter (AFl) with 1:1 conduction may develop, leading to symptoms of presyncope or syncope, particularly during exercise.51 In a cohort of 161 DM1 patients, only 4 of whom exhibited cardiac symptoms, 17% developed AF/AFl during follow-up of 5 ± 4 years.51

Ventricular arrhythmia and SCD

The observation that a portion of DM1 patients still died suddenly despite cardiac pacing raised the suspicion that ventricular tachyarrhythmias underlie this residual risk.58 In a study by Lazarus et al40 on the efficacy of pacemaker implantation for prevention of SCD, 4 such events (2 nonarrhythmic based on postmortem device interrogation) occurred in a population of 49 device recipients.

A multitude of studies sought to determine predictors for SCD; however, only 2 studies provided

Conclusion

Despite significant progress in the understanding and advances in extracardiac support of DM1, gaps in our knowledge of cardiac management and risk stratification remain. Moreover, the majority of available data largely relies on observational data, rendering clinical management prone to wide variation. This could be partly overcome by consolidating care of such patients in tertiary centers with the capacity to provide integrated cardiomyopathy and arrhythmia input for neuromuscular patients.

References (74)

  • J.K. Perloff et al.

    Cardiac involvement in myotonic muscular dystrophy (Steinert's disease): a prospective study of 25 patients

    Am J Cardiol

    (1984)
  • A. Lazarus et al.

    Long-term follow-up of arrhythmias in patients with myotonic dystrophy treated by pacing: a multicenter diagnostic pacemaker study

    J Am Coll Cardiol

    (2002)
  • V. Laurent et al.

    Mortality in myotonic dystrophy patients in the area of prophylactic pacing devices

    Int J Cardiol

    (2011)
  • A. Creta et al.

    A normal electrocardiogram does not exclude infra-hisian conduction disease in patients with myotonic dystrophy type 1

    JACC Clin Electrophysiol

    (2021)
  • H. Petri et al.

    High prevalence of cardiac involvement in patients with myotonic dystrophy type 1: a cross-sectional study

    Int J Cardiol

    (2014)
  • C. Stollberger et al.

    Implantable loop recorders in myotonic dystrophy 1

    Int J Cardiol

    (2011)
  • K. Wahbi et al.

    Atrial flutter in myotonic dystrophy type 1: patient characteristics and clinical outcome

    Neuromuscul Disord

    (2016)
  • A. Lazarus et al.

    Relationship between cardiac arrhythmias and sleep apnoea in permanently paced patients with type I myotonic dystrophy

    Neuromuscul Disord

    (2007)
  • L.E. Grigg et al.

    Ventricular tachycardia and sudden death in myotonic dystrophy: clinical, electrophysiologic and pathologic features

    J Am Coll Cardiol

    (1985)
  • J.P. Daubert et al.

    Inappropriate implantable cardioverter-defibrillator shocks in MADIT II: frequency, mechanisms, predictors, and survival impact

    J Am Coll Cardiol

    (2008)
  • A. Cardona et al.

    Myocardial fibrosis by late gadolinium enhancement cardiovascular magnetic resonance in myotonic muscular dystrophy type 1: highly prevalent but not associated with surface conduction abnormality

    J Cardiovasc Magn Reson

    (2019)
  • R. Valaperta et al.

    Cardiac involvement in myotonic dystrophy: the role of troponins and N-terminal pro B-type natriuretic peptide

    Atherosclerosis

    (2017)
  • M. Facenda-Lorenzo et al.

    Cardiac manifestations in myotonic dystrophy type 1 patients followed using a standard protocol in a specialized unit

    Rev Esp Cardiol (Engl Ed)

    (2013)
  • L. Wood et al.

    The UK Myotonic Dystrophy Patient Registry: facilitating and accelerating clinical research

    J Neurol

    (2017)
  • E.J. Kamsteeg et al.

    Best practice guidelines and recommendations on the molecular diagnosis of myotonic dystrophy types 1 and 2

    Eur J Hum Genet

    (2012)
  • G. Meola

    Clinical aspects, molecular pathomechanisms and management of myotonic dystrophies

    Acta Myol

    (2013)
  • J. Mathieu et al.

    A 10-year study of mortality in a cohort of patients with myotonic dystrophy

    Neurology

    (1999)
  • N.E. Johnson et al.

    Relative risks for comorbidities associated with myotonic dystrophy: a population-based analysis

    Muscle Nerve

    (2015)
  • M. Lund et al.

    Cardiac involvement in myotonic dystrophy: a nationwide cohort study

    Eur Heart J

    (2014)
  • E. Lagrue et al.

    A large multicenter study of pediatric myotonic dystrophy type 1 for evidence-based management

    Neurology

    (2019)
  • D. Bhakta et al.

    Increased mortality with left ventricular systolic dysfunction and heart failure in adults with myotonic dystrophy type 1

    Am Heart J

    (2010)
  • D.G. Wansink et al.

    Alternative splicing controls myotonic dystrophy protein kinase structure, enzymatic activity, and subcellular localization

    Mol Cell Biol

    (2003)
  • E.W. Bush et al.

    Myotonic dystrophy protein kinase domains mediate localization, oligomerization, novel catalytic activity, and autoinhibition

    Biochemistry

    (2000)
  • G. Meola et al.

    Myotonic dystrophy type 2: an update on clinical aspects, genetic and pathomolecular mechanism

    J Neuromuscul Dis

    (2015)
  • F. Freyermuth et al.

    Splicing misregulation of SCN5A contributes to cardiac-conduction delay and heart arrhythmia in myotonic dystrophy

    Nat Commun

    (2016)
  • P.D. Pang et al.

    CRISPR-mediated expression of the fetal Scn5a isoform in adult mice causes conduction defects and arrhythmias

    J Am Heart Assoc

    (2018)
  • L. De Ambroggi et al.

    Cardiac involvement in patients with myotonic dystrophy: characteristic features of magnetic resonance imaging

    Eur Heart J Jul

    (1995)
  • Cited by (1)

    Funding Sources: The authors have no funding sources to disclose. Disclosures: The authors have no conflicts of interest to disclose.

    View full text