Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Prince J. Kannankeril is active.

Publication


Featured researches published by Prince J. Kannankeril.


Heart Rhythm | 2013

HRS/EHRA/APHRS Expert Consensus Statement on the Diagnosis and Management of Patients with Inherited Primary Arrhythmia Syndromes: Document endorsed by HRS, EHRA, and APHRS in May 2013 and by ACCF, AHA, PACES, and AEPC in June 2013.

Silvia G. Priori; Arthur A.M. Wilde; Minoru Horie; Yongkeun Cho; Elijah R. Behr; Charles I. Berul; Nico A. Blom; Josep Brugada; Chern En Chiang; Heikki V. Huikuri; Prince J. Kannankeril; Andrew D. Krahn; Antoine Leenhardt; Arthur J. Moss; Peter J. Schwartz; Wataru Shimizu; Gordon F. Tomaselli; Cynthia Tracy

Developed in partnership with the Heart Rhythm Society (HRS), the European Heart Rhythm Association (EHRA), a registered branch of the European Society of Cardiology, and the Asia Pacific Heart Rhythm Society (APHRS); and in collaboration with the American College of Cardiology Foundation (ACCF), the American Heart Association (AHA), the Pediatric and Congenital Electrophysiology Society (PACES) and the Association for European Pediatric and Congenital Cardiology (AEPC).


Journal of the American College of Cardiology | 2008

Assessment of autonomic function in cardiovascular disease: physiological basis and prognostic implications.

Marc K. Lahiri; Prince J. Kannankeril; Jeffrey J. Goldberger

Certain abnormalities of autonomic function in the setting of structural cardiovascular disease have been associated with an adverse prognosis. Various markers of autonomic activity have received increased attention as methods for identifying patients at risk for sudden death. Both the sympathetic and the parasympathetic limbs can be characterized by tonic levels of activity, which are modulated by, and respond reflexively to, physiological changes. Heart rate provides an index of the net effects of autonomic tone on the sinus node, and carries prognostic significance. Heart rate variability, though related to heart rate, assesses modulation of autonomic control of heart rate and carries additional prognostic information, which in some cases is more powerful than heart rate alone. Heart rate recovery after exercise represents the changes in autonomic tone that occur immediately after cessation of exercise. This index has also been shown to have prognostic significance. Autonomic evaluation during exercise and recovery may be important prognostically, because these are high-risk periods for sudden death, and the autonomic changes that occur with exercise could modulate this high risk. These markers provide related, but not redundant information about different aspects of autonomic effects on the sinus node.


Pharmacological Reviews | 2010

Drug-Induced Long QT Syndrome

Prince J. Kannankeril; Dan M. Roden; Dawood Darbar

The drug-induced long QT syndrome is a distinct clinical entity that has evolved from an electrophysiologic curiosity to a centerpiece in drug regulation and development. This evolution reflects an increasing recognition that a rare adverse drug effect can profoundly upset the balance between benefit and risk that goes into the prescription of a drug by an individual practitioner as well as the approval of a new drug entity by a regulatory agency. This review will outline how defining the central mechanism, block of the cardiac delayed-rectifier potassium current IKr, has contributed to defining risk in patients and in populations. Models for studying risk, and understanding the way in which clinical risk factors modulate cardiac repolarization at the molecular level are discussed. Finally, the role of genetic variants in modulating risk is described.


Circulation | 2008

Cardiac Sodium Channel (SCN5A) Variants Associated with Atrial Fibrillation

Dawood Darbar; Prince J. Kannankeril; Brian S. Donahue; Gayle Kucera; Tanya Stubblefield; Jonathan L. Haines; Alfred L. George; Dan M. Roden

Background— Genetic studies have identified ion channel gene variants in families segregating atrial fibrillation (AF), the most common arrhythmia in clinical practice. Here, we tested the hypothesis that vulnerability to AF is associated with variation in SCN5A, the gene encoding the cardiac sodium channel. Methods and Results— We resequenced the entire SCN5A coding region in 375 subjects with either lone AF (n=118) or AF associated with heart disease (n=257). Controls (n=360) from the same population were then genotyped for the presence of mutations or rare variants identified in the AF cases. In 10 probands (2.7%), 8 novel variants not found in the control population (0%; P=0.001) were identified. All variants affect highly conserved residues in the SCN5A protein. In 6 families with >1 affected member, the novel variant cosegregated with AF. We also identified 11 rare missense variants in 12 probands (3.2%) that have previously been associated with inherited arrhythmia syndromes (eg, congenital long-QT syndrome and Brugada syndrome). Conclusions— Mutations or rare variants in SCN5A may predispose patients with or without underlying heart disease to AF. The findings of the present study expand the clinical spectrum of disorders of the cardiac sodium channel to include AF and represent important progress toward molecular phenotyping and directed rather than empirical therapy for this common arrhythmia.


Journal of the American College of Cardiology | 2011

Flecainide therapy reduces exercise-induced ventricular arrhythmias in patients with catecholaminergic polymorphic ventricular tachycardia

Christian van der Werf; Prince J. Kannankeril; Frederic Sacher; Andrew D. Krahn; Sami Viskin; Antoine Leenhardt; Wataru Shimizu; Naokata Sumitomo; Frank A. Fish; Zahurul A. Bhuiyan; Albert R. Willems; Maurits J. van der Veen; Hiroshi Watanabe; Julien Laborderie; Michel Haïssaguerre; Björn C. Knollmann; Arthur A.M. Wilde

OBJECTIVES This study evaluated the efficacy and safety of flecainide in addition to conventional drug therapy in patients with catecholaminergic polymorphic ventricular tachycardia (CPVT). BACKGROUND CPVT is an inherited arrhythmia syndrome caused by gene mutations that destabilize cardiac ryanodine receptor Ca(2+) release channels. Sudden cardiac death is incompletely prevented by conventional drug therapy with β-blockers with or without Ca(2+) channel blockers. The antiarrhythmic agent flecainide directly targets the molecular defect in CPVT by inhibiting premature Ca(2+) release and triggered beats in vitro. METHODS We collected data from every consecutive genotype-positive CPVT patient started on flecainide at 8 international centers before December 2009. The primary outcome measure was the reduction of ventricular arrhythmias during exercise testing. RESULTS Thirty-three patients received flecainide because of exercise-induced ventricular arrhythmias despite conventional (for different reasons, not always optimal) therapy (median age 25 years; range 7 to 68 years; 73% female). Exercise tests comparing flecainide in addition to conventional therapy with conventional therapy alone were available for 29 patients. Twenty-two patients (76%) had either partial (n = 8) or complete (n = 14) suppression of exercise-induced ventricular arrhythmias with flecainide (p < 0.001). No patient experienced worsening of exercise-induced ventricular arrhythmias. The median daily flecainide dose in responders was 150 mg (range 100 to 300 mg). During a median follow-up of 20 months (range 12 to 40 months), 1 patient experienced implantable cardioverter-defibrillator shocks for polymorphic ventricular arrhythmias, which were associated with a low serum flecainide level. In 1 patient, flecainide successfully suppressed exercise-induced ventricular arrhythmias for 29 years. CONCLUSIONS Flecainide reduced exercise-induced ventricular arrhythmias in patients with CPVT not controlled by conventional drug therapy.


Heart Rhythm | 2013

Executive summary: HRS/EHRA/APHRS expert consensus statement on the diagnosis and management of patients with inherited primary arrhythmia syndromes.

Silvia G. Priori; Arthur A.M. Wilde; Minoru Horie; Yongkeun Cho; Elijah R. Behr; Charles I. Berul; Nico A. Blom; Josep Brugada; Chern En Chiang; Heikki V. Huikuri; Prince J. Kannankeril; Andrew D. Krahn; Antoine Leenhardt; Arthur J. Moss; Peter J. Schwartz; Wataru Shimizu; Gordon F. Tomaselli; Cynthia Tracy

and Management of Patients with Inherited Primary Arrhythmia Syndromes Silvia G. Priori, MD, PhD, (HRS Chairperson), Arthur A. Wilde, MD, PhD, (EHRA Chairperson), Minoru Horie, MD, PhD, (APHRS Chairperson), Yongkeun Cho, MD, PhD, (APHRS Chairperson), Elijah R. Behr, MA, MBBS, MD, FRCP, Charles Berul, MD, FHRS, CCDS, Nico Blom, MD, PhD*, Josep Brugada, MD, PhD, Chern-En Chiang, MD, PhD, Heikki Huikuri, MD, Prince Kannankeril, MD, Andrew Krahn, MD, FHRS, Antoine Leenhardt, MD, Arthur Moss, MD, Peter J. Schwartz, MD, Wataru Shimizu, MD, PhD, Gordon Tomaselli, MD, FHRS, Cynthia Tracy, MD From the Maugeri Foundation IRCCS, Pavia, Italy, Department of Molecular Medicine, University of Pavia, Pavia, Italy and New York University, New York, New York, Department of Cardiology, Academic Medical Centre, Amsterdam, Netherlands, Princess Al-Jawhara Al-Brahim Centre of Excellence in Research of Hereditary Disorders, Jeddah, Kingdom of Saudi Arabia, Shiga University of Medical Sciences, Otsu, Japan, Kyungpook National University Hospital, Daegu, South Korea, St. Georges University of London, United Kingdom, Children’s National Medical Center, Washington, DC, United States, Academical Medical Center, Amsterdam, Leiden University Medical Center, Leiden, Netherlands, University of Barcelona, Barcelona, Spain, Taipei Veteran’s General Hospital, Taipei, Taiwan, Oulu University Central Hospital, Oulu, Finland, Vanderbilt Children’s Hospital, Nashville, Tennessee, United States, Sauder Family and Heart and Stroke Foundation University of British Columbia, British Columbia, Canada, Bichat University Hospital, Paris, France, University of Rochester Medical Center, Rochester, New York, United States, Department of Molecular Medicine, University of Pavia, Pavia, Italy, Nippon Medical School, Tokyo, Japan, Johns Hopkins University, Baltimore, Maryland, United States, and George Washington University Medical Center, Washington, DC, United States.


Current Opinion in Cardiology | 2007

Drug-induced long QT and torsade de pointes: recent advances.

Prince J. Kannankeril; Dan M. Roden

Purpose of review A wide array of drugs can cause marked QT prolongation with the associated risk of torsade de pointes. The large number of drugs with this potential, the correspondingly large number of patients exposed to such drugs, and the potentially fatal outcome make drug-induced long QT syndrome an important public health problem. This review focuses on mechanisms underlying QT prolongation and proarrhythmia, risk factors, including the role of genetic variants, and the unifying framework of reduced repolarization reserve. Recent findings While most drugs that prolong the QT block a specific potassium channel, novel mechanisms altering protein trafficking have been discovered. The progression to torsade de pointes may be less related to degree of QT prolongation than to drug effects on transmural dispersion or variability of repolarization. Our understanding of certain predisposing risk factors has been further refined. Summary Ongoing research continues to elucidate the mechanisms underlying drug-induced long QT syndrome. Importantly, studies are establishing improved predictors of risk for progression to torsade de pointes, in addition to the degree of QT prolongation, which is an imperfect predictor. Nonetheless, drug-induced long QT syndrome and torsade de pointes pose unique challenges for clinicians, researchers, drug-development programs, and regulatory agencies.


Journal of Arrhythmia | 2014

HRS/EHRA/APHRS Expert Consensus Statement on the Diagnosis and Management of Patients with Inherited Primary Arrhythmia Syndromes

Silvia G. Priori; Arthur A.M. Wilde; Minoru Horie; Yongkeun Cho; Elijah R. Behr; Charles I. Berul; Nico A. Blom; Josep Brugada; Chern En Chiang; Heikki V. Huikuri; Prince J. Kannankeril; Andrew D. Krahn; Antoine Leenhardt; Arthur J. Moss; Peter J. Schwartz; Wataru Shimizu; Gordon F. Tomaselli; Cynthia Tracy

Developed in partnership with the Heart Rhythm Society (HRS), the European Heart Rhythm Association (EHRA), a registered branch of the European Society of Cardiology, and the Asia Pacific Heart Rhythm Society (APHRS); and in collaboration with the American College of Cardiology Foundation (ACCF), the American Heart Association (AHA), the Pediatric and Congenital Electrophysiology Society (PACES) and the Association for European Pediatric and Congenital Cardiology (AEPC).


European Heart Journal | 2008

Large scale replication and meta-analysis of variants on chromosome 4q25 associated with atrial fibrillation.

Stefan Kääb; Dawood Darbar; Charlotte van Noord; Josée Dupuis; Arne Pfeufer; Christopher Newton-Cheh; Renate B. Schnabel; Seiko Makino; Moritz F. Sinner; Prince J. Kannankeril; Britt M. Beckmann; Subbarao Choudry; Brian S. Donahue; Jan Heeringa; Siegfried Perz; Kathryn L. Lunetta; Martin G. Larson; Daniel Levy; Calum A. MacRae; Jeremy N. Ruskin; Annette Wacker; Albert Schömig; H.-Erich Wichmann; Gerhard Steinbeck; Thomas Meitinger; André G. Uitterlinden; Jacqueline C. M. Witteman; Dan M. Roden; Emelia J. Benjamin; Patrick T. Ellinor

AIMS A recent genome-wide association study identified a haplotype block on chromosome 4q25 associated with atrial fibrillation (AF). We sought to replicate this association in four independent cohorts. METHODS AND RESULTS The Framingham Heart Study and Rotterdam Study are community-based longitudinal studies. The Vanderbilt AF Registry and German AF Network (AFNet) are case-control studies. Participants with AF (n = 3508) were more likely to be male and were older than referent participants (n = 12 173; Framingham 82 +/- 10 vs. 71 +/- 13 years; Rotterdam 73 +/- 8 vs. 69 +/- 9 years; Vanderbilt 54 +/- 14 vs. 57 +/- 14 years; AFNet 62 +/- 12 vs. 49 +/- 14 years). Single nucleotide polymorphism (SNP) rs2200733 was associated with AF in all four cohorts, with odds ratios (ORs) ranging from 1.37 in Rotterdam [95% confidence interval (CI) 1.18-1.59; P = 3.1 x 10(-5)] to 2.52 in AFNet (95% CI 2.22-2.8; P = 1.8 x 10(-49)). There also was a significant association between AF and rs10033464 in Framingham (OR 1.34; 95% CI 1.03-1.75; P = 0.031) and AFNet (OR 1.30; 95% CI 1.13-1.51; P = 0.0002), but not Vanderbilt (OR 1.16; 95% CI 0.86-1.56; P = 0.33). A meta-analysis of the current and prior AF studies revealed an OR of 1.90 (95% CI 1.60-2.26; P = 3.3 x 10(-13)) for rs2200733 and of 1.36 (95% CI 1.26-1.47; P = 6.7 x 10(-15)) for rs10033464. CONCLUSION The non-coding SNPs rs2200733 and rs10033464 are strongly associated with AF in four cohorts of European descent. These results confirm the significant relations between AF and intergenic variants on chromosome 4.


Circulation-arrhythmia and Electrophysiology | 2009

Mutations in sodium channel β1- and β2-subunits associated with atrial fibrillation.

Hiroshi Watanabe; Dawood Darbar; Daniel W. Kaiser; Kim Jiramongkolchai; Sameer S. Chopra; Brian S. Donahue; Prince J. Kannankeril; Dan M. Roden

Background—We and others have reported mutations in the cardiac predominant sodium channel gene SCN5A in patients with atrial fibrillation (AF). We also have reported that SCN1B is associated with Brugada syndrome and isolated cardiac conduction disease. We tested the hypothesis that mutations in the 4 sodium channel β-subunit genes SCN1B–SCN4B contribute to AF susceptibility. Methods and Results—Screening for mutations in the 4 β-subunit genes was performed in 480 patients with AF (118 patients with lone AF and 362 patients with AF and cardiovascular disease) and 548 control subjects (188 ethnically defined anonymized subjects and 360 subjects without AF). The effects of mutant β-subunits on SCN5A mediated currents were studied using electrophysiological studies. We identified 2 nonsynonymous variants in SCN1B (resulting in R85H, D153N) and 2 in SCN2B (R28Q, R28W) in patients with AF. These occur at residues highly conserved across mammals and were absent in control subjects. In 3 of 4 mutation carriers, the ECGs showed saddleback-type ST-segment elevation in the right precordial leads. Transcripts encoding both SCN1B and SCN2B were detected in human atrium and ventricle. In heterologous expression studies using Chinese hamster ovary cells, the mutant β1- or β2-subunits reduced SCN5A-mediated current and altered channel gating compared with coexpression of wild-type subunits. Conclusions—Loss of function mutations in sodium channel β-subunits were identified in patients with AF and were associated with a distinctive ECG phenotype. These findings further support the hypothesis that decreased sodium current enhances AF susceptibility.

Collaboration


Dive into the Prince J. Kannankeril's collaboration.

Top Co-Authors

Avatar

Dan M. Roden

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew H. Smith

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sara L. Van Driest

Vanderbilt University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Minoru Horie

Shiga University of Medical Science

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge