Network


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

Hotspot


Dive into the research topics where Darwin Jeyaraj is active.

Publication


Featured researches published by Darwin Jeyaraj.


Nature | 2012

Circadian rhythms govern cardiac repolarization and arrhythmogenesis.

Darwin Jeyaraj; Saptarsi M. Haldar; Xiaoping Wan; Mark D. McCauley; Juergen Ripperger; Kun Hu; Yuan Lu; Betty L. Eapen; Nikunj Sharma; Eckhard Ficker; Michael J. Cutler; James Gulick; Atsushi Sanbe; Jeffrey Robbins; Sophie Demolombe; Roman V. Kondratov; Steven Shea; Urs Albrecht; Xander H.T. Wehrens; David S. Rosenbaum; Mukesh K. Jain

Sudden cardiac death exhibits diurnal variation in both acquired and hereditary forms of heart disease, but the molecular basis of this variation is unknown. A common mechanism that underlies susceptibility to ventricular arrhythmias is abnormalities in the duration (for example, short or long QT syndromes and heart failure) or pattern (for example, Brugada’s syndrome) of myocardial repolarization. Here we provide molecular evidence that links circadian rhythms to vulnerability in ventricular arrhythmias in mice. Specifically, we show that cardiac ion-channel expression and QT-interval duration (an index of myocardial repolarization) exhibit endogenous circadian rhythmicity under the control of a clock-dependent oscillator, krüppel-like factor 15 (Klf15). Klf15 transcriptionally controls rhythmic expression of Kv channel-interacting protein 2 (KChIP2), a critical subunit required for generating the transient outward potassium current. Deficiency or excess of Klf15 causes loss of rhythmic QT variation, abnormal repolarization and enhanced susceptibility to ventricular arrhythmias. These findings identify circadian transcription of ion channels as a mechanism for cardiac arrhythmogenesis.


Circulation | 2007

Mechanoelectrical Feedback as Novel Mechanism of Cardiac Electrical Remodeling

Darwin Jeyaraj; Lance D. Wilson; Jia Zhong; Chris A. Flask; Jeffrey E. Saffitz; Isabelle Deschênes; Xin Yu; David S. Rosenbaum

Background— Altered electrical activation of the heart by pacing or disease induces profound ventricular electrical remodeling (VER), manifested electrocardiographically as T-wave memory and ultimately as deleterious mechanical remodeling from heterogeneous strain. Although T-wave memory is associated with altered expression of sarcolemmal ion channels, the biophysical mechanisms responsible for triggering remodeling of cardiac ion channels are unknown. Methods and Results— To test the hypothesis that mechanoelectrical feedback triggered by regional strain is a mechanism for VER, dogs (n=6) underwent 4 weeks of ventricular pacing to induce VER. Multisegment transmural optical action potential imaging of left ventricular wedges revealed profound and selective prolongation of action potential duration in late-activated (288±29 ms) compared with early-activated (250±9 ms) myocardial segments (P<0.05), providing the first experimental evidence that amplification of repolarization gradients between segments of left ventricle is the electrophysiological basis for T-wave memory. In vivo tagged magnetic resonance imaging revealed a 2-fold and preferential increase in circumferential strain in late-activated segments of myocardium, which exactly coincided with segments undergoing VER. VER could not be attributed to structural remodeling because it occurred without any histological evidence of cellular hypertrophy. Conclusions— The mechanism responsible for triggering remodeling of ion channel function in VER was locally enhanced circumferential strain. These data suggest a novel mechanoelectrical feedback mechanism for inducing physiological and potentially deleterious electrical heterogeneities in the heart.


Heart Rhythm | 2009

Heart failure enhances susceptibility to arrhythmogenic cardiac alternans

Lance D. Wilson; Darwin Jeyaraj; Xiaoping Wan; Gregory S. Hoeker; Tamer H. Said; Matthew Gittinger; Kenneth R. Laurita; David S. Rosenbaum

BACKGROUND Although heart failure (HF) is closely associated with susceptibility to sudden cardiac death (SCD), the mechanisms linking contractile dysfunction to cardiac electrical instability are poorly understood. Cardiac alternans has also been closely associated with SCD, and has been linked to a mechanism for amplifying electrical heterogeneities in the heart. However, previous studies have focused on alternans in normal rather than failing myocardium. OBJECTIVE This study sought to investigate the hypothesis that HF enhances susceptibility to arrhythmogenic cardiac alternans. METHODS High-resolution transmural optical mapping was performed in canine wedge preparations from normal (n = 8) and HF (n = 8) hearts produced by rapid ventricular pacing. RESULTS HF significantly (P < .004) lowered the heart rate (HR) threshold for action potential duration alternans (APD-ALT) from 236 +/- 25 beats/min to 185 +/- 25 beats/min. In dual optical mapping of action potentials and intracellular Ca experiments (n = 16), HF lowered the HR threshold for Ca-ALT (beat-to-beat alternations of cellular Ca cycling) from 238 +/- 35 to 177 +/- 26 beats/min (P < .005). Importantly: (1) Ca-ALT always either developed at slower HR or simultaneously with APD-ALT in the same cells, and (2) the magnitude of Ca-ALT and APD-ALT were closely correlated (P < .05). HF similarly lowered the HR threshold for Ca-ALT in isolated myocytes under nonalternating action potential clamp, indicating that HF enhances susceptibility to cellular alternans independent of HF-associated changes in repolarization. Importantly, HF significantly (P < .02) lowered the HR threshold for spatially discordant arrhythmogenic alternans (different regions of cells alternating in opposite phase, DIS-ALT). Ventricular fibrillation (VF) was induced in 88% of HF preparations, but only 12% of normal preparations (P < .003) and was uniformly preceded by development of DIS-ALT. CONCLUSION Heart failure increases the susceptibility to arrhythmogenic cardiac alternans, which arises from HF-induced impairment in calcium cycling.


Science Translational Medicine | 2010

Klf15 Deficiency Is a Molecular Link Between Heart Failure and Aortic Aneurysm Formation

Saptarsi M. Haldar; Yuan Lu; Darwin Jeyaraj; Daiji Kawanami; Yingjie Cui; Sam J. Eapen; Caili Hao; Yan Li; Yong-Qiu Doughman; Michiko Watanabe; Koichi Shimizu; Helena Kuivaniemi; Junichi Sadoshima; Kenneth B. Margulies; Thomas P. Cappola; Mukesh K. Jain

The co-occurrence of heart failure and aortic aneurysms may be explained by their common dependence on a transcriptional regulator, which now presents a possible therapeutic target. The aorta should really be considered part of the heart. The elastic walls of this huge vessel expand with each heartbeat, smoothing out the pulses of blood spurting into it from the left ventricle. Some patients with disease in one tend to have disease in the other. By studying genetically engineered mice, Haldar and Lu and colleagues have now identified a transcription factor that seems essential for proper functioning of both organs and that is deficient in patients with cardiomyopathy and aortopathy. This common underlying molecule may help define new approaches for the treatment of patients with co-occurring heart and aortic disease. The authors first noticed that rodents treated with angiotensin II, which stresses the heart and vasculature, causing disease, had very little Kruppel-like factor 15 (KLF15), a key zinc finger transcription factor, in their heart and aorta. The same was true in patients with disease in these areas. Then, by engineering mice that were missing KLF15 from birth, the authors showed that this transcriptional regulator was necessary for proper functioning of the heart and aorta. Without KLF15, mice developed large dilated hearts that could not pump properly and aortas with aneurysms and, in some animals, hematomas. Further investigation in cells from these animals showed that these effects depended on p53 and p300 acetyltransferase likely because KLF15 directly or indirectly inhibits p300-mediated acetylation of p53. As in human diseased tissue, hearts and aorta from Klf15-deficient mice have hyperacetylated p53. Therefore, for some individuals, stress on the heart and aorta may decrease KLF15 concentrations, which sets in motion an undesirable set of cellular signaling cascades that lead to cardiovascular disease. Having a better handle on what these signals are may help us to interfere with their nefarious work. Current therapies for diseases of heart muscle (cardiomyopathy) and aorta (aortopathy) include inhibitors of the renin-angiotensin system, β-adrenergic antagonists, and the statin class of cholesterol-lowering agents. These therapies have limited efficacy, as adverse cardiovascular events continue to occur with some frequency in patients taking these drugs. Although cardiomyopathy and aortopathy can coexist in a number of conditions (for example, Marfan’s syndrome, acromegaly, pregnancy, and aging), pathogenetic molecular links between the two diseases remain poorly understood. We reasoned that identification of common molecular perturbations in these two tissues could point to therapies for both conditions. Here, we show that deficiency of the transcriptional regulator Kruppel-like factor 15 (Klf15) in mice leads to both heart failure and aortic aneurysm formation through a shared molecular mechanism. Klf15 concentrations are markedly reduced in failing human hearts and in human aortic aneurysm tissues. Mice deficient in Klf15 develop heart failure and aortic aneurysms in a p53-dependent and p300 acetyltransferase–dependent fashion. KLF15 activation inhibits p300-mediated acetylation of p53. Conversely, Klf15 deficiency leads to hyperacetylation of p53 in the heart and aorta, a finding that is recapitulated in human tissues. Finally, Klf15-deficient mice are rescued by p53 deletion or p300 inhibition. These findings highlight a molecular perturbation common to the pathobiology of heart failure and aortic aneurysm formation and suggest that manipulation of KLF15 function may be a productive approach to treat these morbid diseases.


Circulation | 2016

Catabolic Defect of Branched-Chain Amino Acids Promotes Heart Failure.

Haipeng Sun; Kristine C. Olson; Chen Gao; Domenick A. Prosdocimo; Meiyi Zhou; Zhihua Wang; Darwin Jeyaraj; Ji Youn Youn; Shuxun Ren; Yunxia Liu; Christoph Rau; Svati H. Shah; Olga Ilkayeva; Wen Jun Gui; Noelle S. William; R. Max Wynn; Christopher B. Newgard; Hua Cai; Xinshu Xiao; David T. Chuang; Paul Christian Schulze; Christopher J. Lynch; Mukesh K. Jain; Yibin Wang

Background— Although metabolic reprogramming is critical in the pathogenesis of heart failure, studies to date have focused principally on fatty acid and glucose metabolism. Contribution of amino acid metabolic regulation in the disease remains understudied. Methods and Results— Transcriptomic and metabolomic analyses were performed in mouse failing heart induced by pressure overload. Suppression of branched-chain amino acid (BCAA) catabolic gene expression along with concomitant tissue accumulation of branched-chain &agr;-keto acids was identified as a significant signature of metabolic reprogramming in mouse failing hearts and validated to be shared in human cardiomyopathy hearts. Molecular and genetic evidence identified the transcription factor Krüppel-like factor 15 as a key upstream regulator of the BCAA catabolic regulation in the heart. Studies using a genetic mouse model revealed that BCAA catabolic defect promoted heart failure associated with induced oxidative stress and metabolic disturbance in response to mechanical overload. Mechanistically, elevated branched-chain &agr;-keto acids directly suppressed respiration and induced superoxide production in isolated mitochondria. Finally, pharmacological enhancement of branched-chain &agr;-keto acid dehydrogenase activity significantly blunted cardiac dysfunction after pressure overload. Conclusions— BCAA catabolic defect is a metabolic hallmark of failing heart resulting from Krüppel-like factor 15–mediated transcriptional reprogramming. BCAA catabolic defect imposes a previously unappreciated significant contribution to heart failure.


Journal of Molecular and Cellular Cardiology | 2010

Krüppel-like factor 4 regulates pressure-induced cardiac hypertrophy

Xudong Liao; Saptarsi M. Haldar; Yuan Lu; Darwin Jeyaraj; Kaavya Paruchuri; Monika Nahori; Yingjie Cui; Klaus H. Kaestner; Mukesh K. Jain

Krüppel-like factors (KLF) are a subfamily of the zinc-finger class of transcriptional regulators that play important roles in diverse cellular processes. While a number of KLFs are expressed in cardiomyocytes, little is known about their specific roles in the heart in vivo. Here, we demonstrate that KLF4 is induced by hypertrophic stimuli in cultured cardiomyocytes and in the mouse heart. Overexpression of KLF4 in neonatal rat ventricular myocytes inhibits three cardinal features of cardiomyocyte hypertrophy: fetal gene expression, protein synthesis, and cell enlargement. Conversely, mice with cardiomyocyte-specific deletion of KLF4 (CM-K4KO) are highly sensitized to transverse aortic constriction (TAC) and exhibit high rates of mortality. CM-K4KO mice that survive TAC display severe pathologic cardiac hypertrophy characterized by increased cardiac mass, depressed LV systolic function, pulmonary congestion, cavity dilation and attenuated LV wall thickening when compared to control genotypes. In addition, CM-K4KO mice develop increased myocardial fibrosis and apoptotic cell death after TAC. Collectively, these studies implicate KLF4 as a novel transcriptional regulator that is indispensible for the hearts response to stress in vivo.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Kruppel-like factor 15 regulates skeletal muscle lipid flux and exercise adaptation

Saptarsi M. Haldar; Darwin Jeyaraj; Priti Anand; Han Zhu; Yuan Lu; Domenick A. Prosdocimo; Betty L. Eapen; Daiji Kawanami; Mitsuharu Okutsu; Leticia Brotto; Hisashi Fujioka; Janos Kerner; Mariana G. Rosca; Owen P. McGuinness; Rod J. Snow; Aaron P. Russell; Anthony N. Gerber; Xiaodong Bai; Zhen Yan; Thomas M. Nosek; Marco Brotto; Charles L. Hoppel; Mukesh K. Jain

The ability of skeletal muscle to enhance lipid utilization during exercise is a form of metabolic plasticity essential for survival. Conversely, metabolic inflexibility in muscle can cause organ dysfunction and disease. Although the transcription factor Kruppel-like factor 15 (KLF15) is an important regulator of glucose and amino acid metabolism, its endogenous role in lipid homeostasis and muscle physiology is unknown. Here we demonstrate that KLF15 is essential for skeletal muscle lipid utilization and physiologic performance. KLF15 directly regulates a broad transcriptional program spanning all major segments of the lipid-flux pathway in muscle. Consequently, Klf15-deficient mice have abnormal lipid and energy flux, excessive reliance on carbohydrate fuels, exaggerated muscle fatigue, and impaired endurance exercise capacity. Elucidation of this heretofore unrecognized role for KLF15 now implicates this factor as a central component of the transcriptional circuitry that coordinates physiologic flux of all three basic cellular nutrients: glucose, amino acids, and lipids.


Trends in Pharmacological Sciences | 2011

Cardiac electrical remodeling in health and disease

Michael J. Cutler; Darwin Jeyaraj; David S. Rosenbaum

Electrical remodeling of the heart takes place in response to both functional (altered electrical activation) and structural (including heart failure and myocardial infarction) stressors. These electrophysiological changes produce a substrate that is prone to malignant ventricular arrhythmias. Understanding the cellular and molecular mechanisms of electrical remodeling is important in elucidating potential therapeutic targets designed to alter maladaptive electrical remodeling. For example, altered patterns of electrical activation lead primarily to electrical remodeling, without significant structural remodeling. By contrast, secondary remodeling arises in response to a structural insult. In this article we review cardiac electrical remodeling (predominantly in the ventricle) with an emphasis on the mechanisms causing these adaptations. These mechanisms suggest novel therapeutic targets for the management or prevention of the most devastating manifestation of heart disease, sudden cardiac death (SCD).


American Journal of Respiratory Cell and Molecular Biology | 2011

Expression Profiling Identifies Klf15 as a Glucocorticoid Target That Regulates Airway Hyperresponsiveness

Kiriko Masuno; Saptarsi M. Haldar; Darwin Jeyaraj; Christina M. Mailloux; Xiaozhu Huang; Rey A. Panettieri; Mukesh K. Jain; Anthony N. Gerber

Glucocorticoids (GCs), which activate GC receptor (GR) signaling and thus modulate gene expression, are widely used to treat asthma. GCs exert their therapeutic effects in part through modulating airway smooth muscle (ASM) structure and function. However, the effects of genes that are regulated by GCs on airway function are not fully understood. We therefore used transcription profiling to study the effects of a potent GC, dexamethasone, on human ASM (HASM) gene expression at 4 and 24 hours. After 24 hours of dexamethasone treatment, nearly 7,500 genes had statistically distinguishable changes in expression; quantitative PCR validation of a 40-gene subset of putative GR-regulated genes in 6 HASM cell lines suggested that the early transcriptional targets of GR signaling are similar in independent HASM lines. Gene ontology analysis implicated GR targets in controlling multiple aspects of ASM function. One GR-regulated gene, the transcription factor, Kruppel-like factor 15 (Klf15), was already known to modulate vascular smooth and cardiac muscle function, but had no known role in the lung. We therefore analyzed the pulmonary phenotype of Klf15(-/-) mice after ovalbumin sensitization and challenge. We found diminished airway responses to acetylcholine in ovalbumin-challenged Klf15(-/-) mice without a significant change in the induction of asthmatic inflammation. In cultured cells, overexpression of Klf15 reduced proliferation of HASM cells, whereas apoptosis in Klf15(-/-) murine ASM cells was increased. Together, these results further characterize the GR-regulated gene network in ASM and establish a novel role for the GR target, Klf15, in modulating airway function.


Circulation-arrhythmia and Electrophysiology | 2011

Enhanced Dispersion of Repolarization Explains Increased Arrhythmogenesis in Severe Versus Therapeutic Hypothermia

Joseph S. Piktel; Darwin Jeyaraj; Tamer H. Said; David S. Rosenbaum; Lance D. Wilson

Background—Hypothermia is proarrhythmic, and, as the use of therapeutic hypothermia (TH) increases, it is critically important to understand the electrophysiological effects of hypothermia on cardiac myocytes and arrhythmia substrates. We tested the hypothesis that hypothermia-enhanced transmural dispersion of repolarization (DOR) is a mechanism of arrhythmogenesis in hypothermia. In addition, we investigated whether the degree of hypothermia, the rate of temperature change, and cooling versus rewarming would alter hypothermia-induced arrhythmia substrates. Methods and Results—Optical action potentials were recorded from cells spanning the transmural wall of canine left ventricular wedge preparations at baseline (36°C), during cooling and during rewarming. Electrophysiological parameters were examined while varying the depth of hypothermia. On cooling to 26°C, DOR increased from 26±4 ms to 93±18 ms (P=0.021); conduction velocity decreased from 35±5 cm/s to 22±5 cm/s (P=0.010). On rewarming to 36°C, DOR remained prolonged, whereas conduction velocity returned to baseline. Conduction block and reentry was observed in all severe hypothermia preparations. Ventricular fibrillation/ventricular tachycardia was seen more during rewarming (4/5) versus cooling (2/6). In TH (n=7), cooling to 32°C mildly increased DOR (31±6 to 50±9, P=0.012), with return to baseline on rewarming and was associated with decreased arrhythmia susceptibility. Increased rate of cooling did not further enhance DOR or arrhythmogenesis. Conclusions—Hypothermia amplifies DOR and is a mechanism for arrhythmogenesis. DOR is directly dependent on the depth of cooling and rewarming. This provides insight into the clinical observation of a low incidence of arrhythmias in TH and has implications for protocols for the clinical application of TH.

Collaboration


Dive into the Darwin Jeyaraj's collaboration.

Top Co-Authors

Avatar

Mukesh K. Jain

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

David S. Rosenbaum

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Saptarsi M. Haldar

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Yuan Lu

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Lance D. Wilson

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Domenick A. Prosdocimo

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Xiaoping Wan

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Yibin Wang

University of California

View shared research outputs
Top Co-Authors

Avatar

Haipeng Sun

Shanghai Jiao Tong University

View shared research outputs
Researchain Logo
Decentralizing Knowledge