Long QT syndrome (LQTS) is characterized by a prolonged QT interval and an increased risk for syncope, seizures, and sudden cardiac arrest. Ventricular fibrillation (VF) stemming from congenital LQTS is a leading identifiable cause for autopsy-negative sudden unexplained death. With a prevalence of 1 in 2500 individuals, LQTS is the most common cardiac channelopathy and has become a model disease for all other cardiac channelopathies. Over the past decade, there have been substantial improvements made in the clinical management of LQTS. However, the level of breakthrough events, the suboptimal side effect profile, and the fact that none of the current therapeutic strategies directy target the pathogenic substrate of LQTS, specifically the dysregulation of cardiac ion channels, motivates the quest for novel therapeutic strategies for LQTS. The development of patient-specific induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) has provided a novel model system to study the pathological substrate of LQTS, as well as test novel therapeutics. With mutations in the KCNQ1-encoded Kv7.1 voltage-gated potassium channel accounting for the pathogenic substrate for the largest portion of gene positive LQTS cases, targeting these defects may hold the greatest therapeutic potential. Due to the dominant-negative characteristics of KCNQ1 mutations along with the evidence that increased levels of mutant protein lead to increased disease severity, reducing the relative amount of mutated protein should attenuate or correct the disease phenotype. Recent advances in gene therapy have underlined the developing role of RNA interference (RNAi) as a potential therapeutic option in cardiac genetic disorders. These studies have all largely used RNAi targeted against a specific pathogenic mutation. For LQTS, this severely limits the clinical utility of a RNAi-based strategy as the vast majority of pathogenic mutations are family specific. However, targeting high frequency synonymous single nucleotide polymorphisms (sSNPs) in KCNQ1, located on the mutated allele, would allow for allele-specific RNAi in a much larger population creating a much broader clinical application. To this end, we plan to develop allele- specific RNAi targeting the two most common KCNQ1 sSNPs and characterize the effect of this RNAi on the LQTS phenotype of patient-specific iPSC-CMs. By validating a SNP-based RNAi approach in LQTS, this method may also hold potential for other genetically mediated channelopathies and cardiomyopathies.

Public Health Relevance

Despite substantial improvements made in the clinical management of Long QT Syndrome (LQTS), a leading identifiable cause for autopsy-negative sudden unexplained death between the ages of 1 and 35, the necessity for new therapeutic strategies is highlighted by breakthrough cardiac events in treated patients, suboptimal side effect profiles of the current therapies, and the fact that none of the current treatment strategie directly target the pathogenic substrate of LQTS. Therefore, the goal of our research is to identify novel therapeutic strategies specifically for LQTS, but whose underlying strategy may have potential for treatment of other cardiac diseases.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Individual Predoctoral NRSA for M.D./Ph.D. Fellowships (ADAMHA) (F30)
Project #
1F30HL127904-01
Application #
8905164
Study Section
Special Emphasis Panel (ZRG1)
Program Officer
Meadows, Tawanna
Project Start
2015-07-01
Project End
2017-06-30
Budget Start
2015-07-01
Budget End
2016-06-30
Support Year
1
Fiscal Year
2015
Total Cost
Indirect Cost
Name
Mayo Clinic, Rochester
Department
Type
DUNS #
006471700
City
Rochester
State
MN
Country
United States
Zip Code
55905
Kapplinger, Jamie D; Erickson, Anders; Asuri, Sirisha et al. (2017) KCNQ1 p.L353L affects splicing and modifies the phenotype in a founder population with long QT syndrome type 1. J Med Genet 54:390-398
Ackerman, Jaeger P; Bartos, Daniel C; Kapplinger, Jamie D et al. (2016) The Promise and Peril of Precision Medicine: Phenotyping Still Matters Most. Mayo Clin Proc :
Murphy, Sinead L; Anderson, Jason H; Kapplinger, Jamie D et al. (2016) Evaluation of the Mayo Clinic Phenotype-Based Genotype Predictor Score in Patients with Clinically Diagnosed Hypertrophic Cardiomyopathy. J Cardiovasc Transl Res 9:153-61
Van Driest, Sara L; Wells, Quinn S; Stallings, Sarah et al. (2016) Association of Arrhythmia-Related Genetic Variants With Phenotypes Documented in Electronic Medical Records. JAMA 315:47-57
Kapplinger, Jamie D; Tseng, Andrew S; Salisbury, Benjamin A et al. (2015) Enhancing the Predictive Power of Mutations in the C-Terminus of the KCNQ1-Encoded Kv7.1 Voltage-Gated Potassium Channel. J Cardiovasc Transl Res 8:187-97
Kapplinger, Jamie D; Giudicessi, John R; Ye, Dan et al. (2015) Enhanced Classification of Brugada Syndrome-Associated and Long-QT Syndrome-Associated Genetic Variants in the SCN5A-Encoded Na(v)1.5 Cardiac Sodium Channel. Circ Cardiovasc Genet 8:582-95