Network


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

Hotspot


Dive into the research topics where Sridharan Rajamani is active.

Publication


Featured researches published by Sridharan Rajamani.


British Journal of Pharmacology | 2006

Drug‐induced long QT syndrome: hERG K+ channel block and disruption of protein trafficking by fluoxetine and norfluoxetine

Sridharan Rajamani; Lee L. Eckhardt; Carmen R. Valdivia; C A Klemens; B M Gillman; Corey L. Anderson; Katherine M. Holzem; Brian P. Delisle; Blake D. Anson; Jonathan C. Makielski; Craig T. January

Fluoxetine (Prozac®) is a widely prescribed drug in adults and children, and it has an active metabolite, norfluoxetine, with a prolonged elimination time. Although uncommon, Prozac causes QT interval prolongation and arrhythmias; a patient who took an overdose of Prozac exhibited a prolonged QT interval (QTc 625 msec). We looked for possible mechanisms underlying this clinical finding by analysing the effects of fluoxetine and norfluoxetine on ion channels in vitro.


Circulation | 2002

Pharmacological Rescue of Human K+ Channel Long-QT2 Mutations Human Ether-a-Go-Go-Related Gene Rescue Without Block

Sridharan Rajamani; Corey L. Anderson; Blake D. Anson; Craig T. January

Background—Defective protein trafficking is a consequence of gene mutations. Human long-QT (LQT) syndrome results from mutations in several genes, including the human ether-a-go-go-related gene (HERG), which encodes a delayed rectifier K+ current. Trafficking-defective mutant HERG protein is a mechanism for reduced delayed rectifier K+ current in LQT2, and high-affinity HERG channel-blocking drugs can result in pharmacological rescue. Methods and Results—We postulated that drug molecules modified to remove high-affinity HERG block may still stabilize mutant proteins in a conformation required for rescue. We tested terfenadine carboxylate (fexofenadine) and terfenadine, structurally similar drugs with markedly different affinities for HERG block, for rescue of trafficking-defective LQT2 mutations. Terfenadine rescued the N470D mutation but blocked the channels. In contrast, fexofenadine rescued N470D with a half-maximal rescue concentration of 177 nmol/L, which is ≈350-fold lower than the half-maximal channel block concentration. The G601S mutation was also rescued without channel block. Conclusions—Pharmacological rescue can occur without channel block. This could represent a new antiarrhythmic paradigm in the treatment of some trafficking-defective LQT2 mutations.


Journal of the American College of Cardiology | 2014

Mutations in SCN10A Are Responsible for a Large Fraction of Cases of Brugada Syndrome

Dan Hu; Hector Barajas-Martinez; Ryan Pfeiffer; Fabio Dezi; Jenna Pfeiffer; Tapan Buch; Matthew J. Betzenhauser; Luiz Belardinelli; Kristopher M. Kahlig; Sridharan Rajamani; Harry J. Deantonio; Robert J. Myerburg; Hiroyuki Ito; Pramod Deshmukh; Mark Marieb; Gi Byoung Nam; Atul Bhatia; Can Hasdemir; Michel Haïssaguerre; Christian Veltmann; Rainer Schimpf; Martin Borggrefe; Sami Viskin; Charles Antzelevitch

BACKGROUND BrS is an inherited sudden cardiac death syndrome. Less than 35% of BrS probands have genetically identified pathogenic variants. Recent evidence has implicated SCN10A, a neuronal sodium channel gene encoding Nav1.8, in the electrical function of the heart. OBJECTIVES The purpose of this study was to test the hypothesis that SCN10A variants contribute to the development of Brugada syndrome (BrS). METHODS Clinical analysis and direct sequencing of BrS susceptibility genes were performed for 150 probands and family members as well as >200 healthy controls. Expression and coimmunoprecipitation studies were performed to functionally characterize the putative pathogenic mutations. RESULTS We identified 17 SCN10A mutations in 25 probands (20 male and 5 female); 23 of the 25 probands (92.0%) displayed overlapping phenotypes. SCN10A mutations were found in 16.7% of BrS probands, approaching our yield for SCN5A mutations (20.1%). Patients with BrS who had SCN10A mutations were more symptomatic and displayed significantly longer PR and QRS intervals compared with SCN10A-negative BrS probands. The majority of mutations localized to the transmembrane-spanning regions. Heterologous coexpression of wild-type (WT) SCN10A with WT-SCN5A in HEK cells caused a near doubling of sodium channel current compared with WT-SCN5A alone. In contrast, coexpression of SCN10A mutants (R14L and R1268Q) with WT-SCN5A caused a 79.4% and 84.4% reduction in sodium channel current, respectively. The coimmunoprecipitation studies provided evidence for the coassociation of Nav1.8 and Nav1.5 in the plasma membrane. CONCLUSIONS Our study identified SCN10A as a major susceptibility gene for BrS, thus greatly enhancing our ability to genotype and risk stratify probands and family members.


Journal of Pharmacology and Experimental Therapeutics | 2013

A Novel, Potent, and Selective Inhibitor of Cardiac Late Sodium Current Suppresses Experimental Arrhythmias

Luiz Belardinelli; Gongxin Liu; Cathy Smith-Maxwell; Wei-Qun Wang; Nesrine El-Bizri; Ryoko Hirakawa; Serge Karpinski; Cindy Hong Li; Lufei Hu; Xiao-Jun Li; William J. Crumb; Lin Wu; Dmitry Koltun; Jeff Zablocki; Lina Yao; Arvinder Dhalla; Sridharan Rajamani; John C. Shryock

Inhibition of cardiac late sodium current (late INa) is a strategy to suppress arrhythmias and sodium-dependent calcium overload associated with myocardial ischemia and heart failure. Current inhibitors of late INa are unselective and can be proarrhythmic. This study introduces GS967 (6-[4-(trifluoromethoxy)phenyl]-3-(trifluoromethyl)-[1,2,4]triazolo[4,3-a]pyridine), a potent and selective inhibitor of late INa, and demonstrates its effectiveness to suppress ventricular arrhythmias. The effects of GS967 on rabbit ventricular myocyte ion channel currents and action potentials were determined. Anti-arrhythmic actions of GS967 were characterized in ex vivo and in vivo rabbit models of reduced repolarization reserve and ischemia. GS967 inhibited Anemonia sulcata toxin II (ATX-II)–induced late INa in ventricular myocytes and isolated hearts with IC50 values of 0.13 and 0.21 µM, respectively. Reduction of peak INa by GS967 was minimal at a holding potential of −120 mV but increased at −80 mV. GS967 did not prolong action potential duration or the QRS interval. GS967 prevented and reversed proarrhythmic effects (afterdepolarizations and torsades de pointes) of the late INa enhancer ATX-II and the IKr inhibitor E-4031 in isolated ventricular myocytes and hearts. GS967 significantly attenuated the proarrhythmic effects of methoxamine+clofilium and suppressed ischemia-induced arrhythmias. GS967 was more potent and effective to reduce late INa and arrhythmias than either flecainide or ranolazine. Results of all studies and assays of binding and activity of GS967 at numerous receptors, transporters, and enzymes indicated that GS967 selectively inhibited late INa. In summary, GS967 selectively suppressed late INa and prevented and/or reduced the incidence of experimentally induced arrhythmias in rabbit myocytes and hearts.


American Journal of Physiology-cell Physiology | 2011

Nav1.5-dependent persistent Na+ influx activates CaMKII in rat ventricular myocytes and N1325S mice

Lina Yao; Peidong Fan; Zhan Jiang; Serge Viatchenko-Karpinski; Yuzhi Wu; Dmytro Kornyeyev; Ryoko Hirakawa; Grant R. Budas; Sridharan Rajamani; John C. Shryock; Luiz Belardinelli

Late Na(+) current (I(NaL)) and Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) are both increased in the diseased heart. Recently, CaMKII was found to phosphorylate the Na(+) channel 1.5 (Na(v)1.5), resulting in enhanced I(NaL). Conversely, an increase of I(NaL) would be expected to cause elevation of intracellular Ca(2+) and activation of CaMKII. However, a relationship between enhancement of I(NaL) and activation of CaMKII has yet to be demonstrated. We investigated whether Na(+) influx via Na(v)1.5 leads to CaMKII activation and explored the functional significance of this pathway. In neonatal rat ventricular myocytes (NRVM), treatment with the I(NaL) activators anemone toxin II (ATX-II) or veratridine increased CaMKII autophosphorylation and increased phosphorylation of CaMKII substrates phospholamban and ryanodine receptor 2. Knockdown of Na(v)1.5 (but not Na(v)1.1 or Na(v)1.2) prevented ATX-II-induced CaMKII phosphorylation, providing evidence for a specific role of Na(v)1.5 in CaMKII activation. In support of this view, CaMKII activity was also increased in hearts of transgenic mice overexpressing a gain-of-function Na(v)1.5 mutant (N(1325)S). The effects of both ATX-II and the N(1325)S mutation were reversed by either I(NaL) inhibition (with ranolazine or tetrodotoxin) or CaMKII inhibition (with KN93 or autocamtide 2-related inhibitory peptide). Furthermore, ATX-II treatment also induced CaMKII-Na(v)1.5 coimmunoprecipitation. The same association between CaMKII and Na(v)1.5 was also found in N(1325)S mice, suggesting a direct protein-protein interaction. Pharmacological inhibitions of either CaMKII or I(NaL) also prevented ATX-II-induced cell death in NRVM and reduced the incidence of polymorphic ventricular tachycardia induced by ATX-II in rat perfused hearts. Taken together, these results suggest that a Na(v)1.5-dependent increase in Na(+) influx leads to activation of CaMKII, which in turn phosphorylates Na(v)1.5, further promoting Na(+) influx. Pharmacological inhibition of either CaMKII or Na(v)1.5 can ameliorate cardiac dysfunction caused by excessive Na(+) influx.


Heart Rhythm | 2009

Use-dependent block of cardiac late Na+ current by ranolazine

Sridharan Rajamani; Nesrine El-Bizri; John C. Shryock; Jonathan C. Makielski; Luiz Belardinelli

BACKGROUND Ranolazine is an antianginal drug that inhibits the cardiac late Na+ current (INa). The selectivity of ranolazine to block late INa relative to peak INa at rapid heart rates has not been determined, but is potentially important to drug efficacy and safety. OBJECTIVE This study sought to quantify use-dependent block (UDB) of cardiac peak and late INa by ranolazine. METHODS Wild-type (WT) and long QT3 mutation R1623Q channels were expressed in HEK293 cells and studied using whole-cell patch-clamp technique. RESULTS Ranolazine (1 to 300 microM caused tonic (0.1 Hz) and UDB (1, 2, and 5 Hz) of WT and R1623Q peak INa. The IC50 values for block of WT and R1623Q peak INa at 0.1, 1, 2, and 5 Hz were 430, 260, 157, and 154 microM, and 95, 77, 37, and 25 microM, respectively. The IC50 values for block of R1623Q late INa at 0.1, 1, 2, and 5 Hz were 7.5, 7.3, 2.2, and 1.9 microM, respectively. Ranolazine (10 microM) caused a hyperpolarizing shift of WT and R1623Q peak INa steady-state inactivation without affecting steady-state activation, suggesting that ranolazine interacts with inactivated states of the channels. Ranolazine (30 microM) significantly slowed the recovery from inactivation of peak INa of both WT and R1623Q and late INa of R1623Q. CONCLUSION Ranolazine slowed recovery of late INa from inactivation and thus caused UDB of late INa. These data suggest that the effect of ranolazine to block late INa may be increased and the selectivity to block late INa relative to peak INa may be retained during tachycardia.


Handbook of experimental pharmacology | 2014

The Role of Late I Na in Development of Cardiac Arrhythmias

Charles Antzelevitch; Vladislav V. Nesterenko; John C. Shryock; Sridharan Rajamani; Yejia Song; Luiz Belardinelli

Late I Na is an integral part of the sodium current, which persists long after the fast-inactivating component. The magnitude of the late I Na is relatively small in all species and in all types of cardiomyocytes as compared with the amplitude of the fast sodium current, but it contributes significantly to the shape and duration of the action potential. This late component had been shown to increase in several acquired or congenital conditions, including hypoxia, oxidative stress, and heart failure, or due to mutations in SCN5A, which encodes the α-subunit of the sodium channel, as well as in channel-interacting proteins, including multiple β subunits and anchoring proteins. Patients with enhanced late I Na exhibit the type-3 long QT syndrome (LQT3) characterized by high propensity for the life-threatening ventricular arrhythmias, such as Torsade de Pointes (TdP), as well as for atrial fibrillation. There are several distinct mechanisms of arrhythmogenesis due to abnormal late I Na, including abnormal automaticity, early and delayed after depolarization-induced triggered activity, and dramatic increase of ventricular dispersion of repolarization. Many local anesthetic and antiarrhythmic agents have a higher potency to block late I Na as compared with fast I Na. Several novel compounds, including ranolazine, GS-458967, and F15845, appear to be the most selective inhibitors of cardiac late I Na reported to date. Selective inhibition of late I Na is expected to be an effective strategy for correcting these acquired and congenital channelopathies.


Heart Rhythm | 2015

Cardiac late Na+ current: Proarrhythmic effects, roles in long QT syndromes, and pathological relationship to CaMKII and oxidative stress

Luiz Belardinelli; Wayne R. Giles; Sridharan Rajamani; Hrayr S. Karagueuzian; John C. Shryock

Myocyte sodium channel current that persists throughout the plateau of the cardiac action potential is referred to as late sodium current (I(Na-L)). The magnitude of I(Na-L) is normally small, but can increase significantly in common acute and chronic pathological settings as a result of inherited and/or acquired Na(+) channelopathies that alter channel opening and closing (ie, gating), location (trafficking), or anchoring and interactions with cytoskeletal proteins. An increase in I(Na-L) reduces repolarization reserve in atrial and ventricular myocytes and prolongs the action potential duration and the QT interval. An enhanced I(Na-L) is a cause of long QT syndrome 3. I(Na-L) may be a cause of afterdepolarizations, triggered arrhythmias, and spontaneous diastolic depolarization-induced automaticity. In addition, enhancement of I(Na-L) increases both the temporal and the spatial dispersion of repolarization in the myocardium and may lead to spatially discordant action potential duration alternans, wavebreak, and reentrant arrhythmias. Positive feedback loops between increases in I(Na-L) and the activity of Ca(2+)/calmodulin-dependent protein kinase II appear to contribute to the genesis of arrhythmias and to certain abnormalities of the ischemic heart. In this review, we discuss some of the more relevant experimental results, clinical findings, and insights from cellular and animal models that highlight the role of I(Na-L) in the genesis of arrhythmias, long QT syndromes, and intracellular Ca(2+) homeostasis.


PLOS ONE | 2012

Simulation and Mechanistic Investigation of the Arrhythmogenic Role of the Late Sodium Current in Human Heart Failure

Beatriz Trenor; Karen Cardona; Juan F. Gomez; Sridharan Rajamani; J.M. Ferrero; Luiz Belardinelli; Javier Saiz

Heart failure constitutes a major public health problem worldwide. The electrophysiological remodeling of failing hearts sets the stage for malignant arrhythmias, in which the role of the late Na+ current (INaL) is relevant and is currently under investigation. In this study we examined the role of INaL in the electrophysiological phenotype of ventricular myocytes, and its proarrhythmic effects in the failing heart. A model for cellular heart failure was proposed using a modified version of Grandi et al. model for human ventricular action potential that incorporates the formulation of INaL. A sensitivity analysis of the model was performed and simulations of the pathological electrical activity of the cell were conducted. The proposed model for the human INaL and the electrophysiological remodeling of myocytes from failing hearts accurately reproduce experimental observations. The sensitivity analysis of the modulation of electrophysiological parameters of myocytes from failing hearts due to ion channels remodeling, revealed a role for INaL in the prolongation of action potential duration (APD), triangulation of the shape of the AP, and changes in Ca2+ transient. A mechanistic investigation of intracellular Na+ accumulation and APD shortening with increasing frequency of stimulation of failing myocytes revealed a role for the Na+/K+ pump, the Na+/Ca2+ exchanger and INaL. The results of the simulations also showed that in failing myocytes, the enhancement of INaL increased the reverse rate-dependent APD prolongation and the probability of initiating early afterdepolarizations. The electrophysiological remodeling of failing hearts and especially the enhancement of the INaL prolong APD and alter Ca2+ transient facilitating the development of early afterdepolarizations. An enhanced INaL appears to be an important contributor to the electrophysiological phenotype and to the dysregulation of [Ca2+]i homeostasis of failing myocytes.


American Journal of Physiology-heart and Circulatory Physiology | 2009

Reduction of repolarization reserve unmasks the proarrhythmic role of endogenous late Na(+) current in the heart.

Lin Wu; Sridharan Rajamani; Hong Li; Craig T. January; John C. Shryock; Luiz Belardinelli

Reduction of repolarization reserve increases the risk of arrhythmia. We hypothesized that inhibition of K(+) current (I(K)) to decrease repolarization reserve would unmask the proarrhythmic role of endogenous, physiological late Na(+) current (late I(Na)). Monophasic action potentials (MAP) and 12-lead electrocardiogram were recorded from female rabbit isolated hearts. To block I(K) and reduce repolarization reserve, E-4031, 4-aminopyridine, and BaCl(2) were used; to block endogenous late I(Na), tetrodotoxin (TTX) and ranolazine were used. E-4031 (1-60 nM) concentration-dependently prolonged MAP duration (MAPD(90)) and increased duration of the T wave from T(peak) to T(end) (T(peak)-T(end)), transmural dispersion of repolarization (TDR), and beat-to-beat variability (BVR) of MAPD(90). E-4031 caused spontaneous and pause-triggered polymorphic ventricular tachycardia [torsade de pointes (TdP)]. In the presence of 60 nM E-4031, TTX (0.6-3 muM) and ranolazine (5-10 muM) shortened MAPD(90), decreased TDR, BVR, and T(peak)-T(end) (n = 9-20, P < 0.01), and abolished episodes of TdP. In hearts treated with BaCl(2) or 4-aminopyridine plus E-4031, TTX (0.6-3 muM) shortened MAPD(90) and decreased T(peak)-T(end). Ranolazine could not reverse the effect of E-4031 to inhibit human ether-a-go-go-related gene (HERG) K(+) current; thus, the reversal by ranolazine of effects of E-4031 was likely due to inhibition of late I(Na) and not to antagonism of the HERG-blocking action of E-4031. We conclude that endogenous, physiological late I(Na) contributes to arrhythmogenesis in hearts with reduced repolarization reserve. Inhibition of this current partially reverses MAPD prolongation and abolishes arrhythmic activity caused by I(K) inhibitors.

Collaboration


Dive into the Sridharan Rajamani's collaboration.

Top Co-Authors

Avatar

Luiz Belardinelli

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Blake D. Anson

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Bruce D. Nearing

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge