Network


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

Hotspot


Dive into the research topics where Jeffrey Brandimarto is active.

Publication


Featured researches published by Jeffrey Brandimarto.


Circulation | 2016

Evidence for Intramyocardial Disruption of Lipid Metabolism and Increased Myocardial Ketone Utilization in Advanced Human Heart Failure.

Kenneth C Bedi; Nathaniel W. Snyder; Jeffrey Brandimarto; Moez Karim Aziz; Clementina Mesaros; Andrew J. Worth; Linda L. Wang; Ali Javaheri; Ian A. Blair; Kenneth B. Margulies; J. Eduardo Rame

Background— The failing human heart is characterized by metabolic abnormalities, but these defects remains incompletely understood. In animal models of heart failure there is a switch from a predominance of fatty acid utilization to the more oxygen-sparing carbohydrate metabolism. Recent studies have reported decreases in myocardial lipid content, but the inclusion of diabetic and nondiabetic patients obscures the distinction of adaptations to metabolic derangements from adaptations to heart failure per se. Methods and Results— We performed both unbiased and targeted myocardial lipid surveys using liquid chromatography-mass spectroscopy in nondiabetic, lean, predominantly nonischemic, advanced heart failure patients at the time of heart transplantation or left ventricular assist device implantation. We identified significantly decreased concentrations of the majority of myocardial lipid intermediates, including long-chain acylcarnitines, the primary subset of energetic lipid substrate for mitochondrial fatty acid oxidation. We report for the first time significantly reduced levels of intermediate and anaplerotic acyl-coenzyme A (CoA) species incorporated into the Krebs cycle, whereas the myocardial concentration of acetyl-CoA was significantly increased in end-stage heart failure. In contrast, we observed an increased abundance of ketogenic &bgr;-hydroxybutyryl-CoA, in association with increased myocardial utilization of &bgr;-hydroxybutyrate. We observed a significant increase in the expression of the gene encoding succinyl-CoA:3-oxoacid-CoA transferase, the rate-limiting enzyme for myocardial oxidation of &bgr;-hydroxybutyrate and acetoacetate. Conclusions— These findings indicate increased ketone utilization in the severely failing human heart independent of diabetes mellitus, and they support the role of ketone bodies as an alternative fuel and myocardial ketone oxidation as a key metabolic adaptation in the failing human heart.


Genomics | 2015

RNA-Seq identifies novel myocardial gene expression signatures of heart failure.

Yichuan Liu; Michael Morley; Jeffrey Brandimarto; Sridhar Hannenhalli; Yu Hu; Euan A. Ashley; W.H. Wilson Tang; Christine S. Moravec; Kenneth B. Margulies; Thomas P. Cappola; Mingyao Li

Heart failure is a complex clinical syndrome and has become the most common reason for adult hospitalization in developed countries. Two subtypes of heart failure, ischemic heart disease (ISCH) and dilated cardiomyopathy (DCM), have been studied using microarray platforms. However, microarray has limited resolution. Here we applied RNA sequencing (RNA-Seq) to identify gene signatures for heart failure from six individuals, including three controls, one ISCH and two DCM patients. Using genes identified from this small RNA-Seq dataset, we were able to accurately classify heart failure status in a much larger set of 313 individuals. The identified genes significantly overlapped with genes identified via genome-wide association studies for cardiometabolic traits and the promoters of those genes were enriched for binding sites for transcriptions factors. Our results indicate that it is possible to use RNA-Seq to classify disease status for complex diseases such as heart failure using an extremely small training dataset.


American Journal of Human Genetics | 2014

An Enhancer Polymorphism at the Cardiomyocyte Intercalated Disc Protein NOS1AP Locus Is a Major Regulator of the QT Interval

Ashish Kapoor; Rajesh B. Sekar; Nancy F. Hansen; Karen Fox-Talbot; Michael Morley; Vasyl Pihur; Sumantra Chatterjee; Jeffrey Brandimarto; Christine S. Moravec; Sara L. Pulit; Arne Pfeufer; Jim Mullikin; Mark T. Ross; Eric D. Green; David R. Bentley; Christopher Newton-Cheh; Eric Boerwinkle; Gordon F. Tomaselli; Thomas P. Cappola; Dan E. Arking; Marc K. Halushka; Aravinda Chakravarti

QT interval variation is assumed to arise from variation in repolarization as evidenced from rare Na- and K-channel mutations in Mendelian QT prolongation syndromes. However, in the general population, common noncoding variants at a chromosome 1q locus are the most common genetic regulators of QT interval variation. In this study, we use multiple human genetic, molecular genetic, and cellular assays to identify a functional variant underlying trait association: a noncoding polymorphism (rs7539120) that maps within an enhancer of NOS1AP and affects cardiac function by increasing NOS1AP transcript expression. We further localized NOS1AP to cardiomyocyte intercalated discs (IDs) and demonstrate that overexpression of NOS1AP in cardiomyocytes leads to altered cellular electrophysiology. We advance the hypothesis that NOS1AP affects cardiac electrical conductance and coupling and thereby regulates the QT interval through propagation defects. As further evidence of an important role for propagation variation affecting QT interval in humans, we show that common polymorphisms mapping near a specific set of 170 genes encoding ID proteins are significantly enriched for association with the QT interval, as compared to genome-wide markers. These results suggest that focused studies of proteins within the cardiomyocyte ID are likely to provide insights into QT prolongation and its associated disorders.


Circulation Research | 2017

Pharmacokinetics and Pharmacodynamics of Inorganic Nitrate in Heart Failure With Preserved Ejection Fraction

Payman Zamani; Victor X Tan; Haideliza Soto-Calderon; Melissa Beraun; Jeffrey Brandimarto; Lien Trieu; Swapna Varakantam; Paschalis-Thomas Doulias; Raymond R. Townsend; Jesse Chittams; Kenneth B. Margulies; Thomas P. Cappola; David C. Poole; Harry Ischiropoulos; Julio A. Chirinos

Rationale: Nitrate-rich beetroot juice has been shown to improve exercise capacity in heart failure with preserved ejection fraction, but studies using pharmacological preparations of inorganic nitrate are lacking. Objectives: To determine (1) the dose–response effect of potassium nitrate (KNO3) on exercise capacity; (2) the population-specific pharmacokinetic and safety profile of KNO3 in heart failure with preserved ejection fraction. Methods and Results: We randomized 12 subjects with heart failure with preserved ejection fraction to oral KNO3 (n=9) or potassium chloride (n=3). Subjects received 6 mmol twice daily during week 1, followed by 6 mmol thrice daily during week 2. Supine cycle ergometry was performed at baseline (visit 1) and after each week (visits 2 and 3). Quality of life was assessed with the Kansas City Cardiomyopathy Questionnaire. The primary efficacy outcome, peak O2-uptake, did not significantly improve (P=0.13). Exploratory outcomes included exercise duration and quality of life. Exercise duration increased significantly with KNO3 (visit 1: 9.87, 95% confidence interval [CI] 9.31–10.43 minutes; visit 2: 10.73, 95% CI 10.13–11.33 minute; visit 3: 11.61, 95% CI 11.05–12.17 minutes; P=0.002). Improvements in the Kansas City Cardiomyopathy Questionnaire total symptom (visit 1: 58.0, 95% CI 52.5–63.5; visit 2: 66.8, 95% CI 61.3–72.3; visit 3: 70.8, 95% CI 65.3–76.3; P=0.016) and functional status scores (visit 1: 62.2, 95% CI 58.5–66.0; visit 2: 68.6, 95% CI 64.9–72.3; visit 3: 71.1, 95% CI 67.3–74.8; P=0.01) were seen after KNO3. Pronounced elevations in trough levels of nitric oxide metabolites occurred with KNO3 (visit 2: 199.5, 95% CI 98.7–300.2 &mgr;mol/L; visit 3: 471.8, 95% CI 377.8–565.8 &mgr;mol/L) versus baseline (visit 1: 38.0, 95% CI 0.00–132.0 &mgr;mol/L; P<0.001). KNO3 did not lead to clinically significant hypotension or methemoglobinemia. After 6 mmol of KNO3, systolic blood pressure was reduced by a maximum of 17.9 (95% CI −28.3 to −7.6) mm Hg 3.75 hours later. Peak nitric oxide metabolites concentrations were 259.3 (95% CI 176.2–342.4) &mgr;mol/L 3.5 hours after ingestion, and the median half-life was 73.0 (interquartile range 33.4–232.0) minutes. Conclusions: KNO3 is potentially well tolerated and improves exercise duration and quality of life in heart failure with preserved ejection fraction. This study reinforces the efficacy of KNO3 and suggests that larger randomized trials are warranted. Clinical Trial Registration: URL: http://www.clinicaltrials.gov. Unique identifier: NCT02256345


Journal of Cardiac Failure | 2016

Prognostic Value of Galectin-3 for Adverse Outcomes in Chronic Heart Failure

Benjamin French; Le Wang; Bonnie Ky; Jeffrey Brandimarto; Anupam Basuray; James C. Fang; Nancy K. Sweitzer; Thomas P. Cappola

BACKGROUND Clinical studies have suggested the prognostic value of galectin-3, a marker of fibrosis, in chronic heart failure. However, the specific role of galectin-3, compared with established biomarkers, remains uncertain. METHODS AND RESULTS The Penn Heart Failure Study was an ambulatory heart failure cohort that included 1385 participants with reduced (1141), preserved (106), and recovered (138) left ventricular ejection fraction (LVEF). Cox regression models determined the association between galectin-3 and risk of all-cause mortality, cardiac transplantation, or placement of a ventricular assist device. Receiver operating characteristic curves compared the prognostic accuracy of galectin-3, high-sensitivity soluble Toll-like receptor 2 (ST2), troponin I, and B-type natriuretic peptide (BNP) at 1 and 5 years. Higher galectin-3 levels were associated with an increased risk of adverse events (adjusted hazard ratio of 1.96 for each doubling in galectin-3; P < .001). This association was most pronounced among participants with preserved LVEF (adjusted hazard ratio 3.30; P < .001). At 5 years, galectin-3 was the most accurate discriminator of risk among participants with preserved LVEF (area under the curve 0.782; P = .81 vs high-sensitivity ST2; P = .029 vs troponin I; P = .35 vs BNP). BNP was most accurate among participants with reduced and recovered LVEF (areas under the curves 0.716 and 0.728, respectively). CONCLUSIONS Galectin-3 could have prognostic value for long-term events among patients with heart failure and preserved ejection fraction.


Journal of the American College of Cardiology | 2015

Systems Genomics Identifies a Key Role for Hypocretin/Orexin Receptor-2 in Human Heart Failure.

Marco V Perez; Aleksandra Pavlovic; Ching Shang; Matthew T. Wheeler; Clint L. Miller; Jing Liu; Frederick E. Dewey; Stephen Pan; Porama Thanaporn; Devin Absher; Jeffrey Brandimarto; Heidi Salisbury; Khin Chan; Rupak Mukherjee; Roda P. Konadhode; Richard M. Myers; Daniel Sedehi; Thomas E. Scammell; Thomas Quertermous; Thomas P. Cappola; Euan A. Ashley

BACKGROUND The genetic determinants of heart failure (HF) and response to medical therapy remain unknown. We hypothesized that identifying genetic variants of HF that associate with response to medical therapy would elucidate the genetic basis of cardiac function. OBJECTIVES This study sought to identify genetic variations associated with response to HF therapy. METHODS This study compared extremes of response to medical therapy in 866 HF patients using a genome-wide approach that informed the systems-based design of a customized single nucleotide variant array. The effect of genotype on gene expression was measured using allele-specific luciferase reporter assays. Candidate gene transcription-deficient mice underwent echocardiography and treadmill exercise. The ability of the target gene agonist to rescue mice from chemically-induced HF was assessed with echocardiography. RESULTS Of 866 HF patients, 136 had an ejection fraction improvement of 20% attributed to resynchronization (n = 83), revascularization (n = 7), tachycardia resolution (n = 2), alcohol cessation (n = 1), or medications (n = 43). Those with the minor allele for rs7767652, upstream of hypocretin (orexin) receptor-2 (HCRTR2), were less likely to have improved left ventricular function (odds ratio: 0.40 per minor allele; p = 3.29 × 10(-5)). In a replication cohort of 798 patients, those with a minor allele for rs7767652 had a lower prevalence of ejection fraction >35% (odds ratio: 0.769 per minor allele; p = 0.021). In an HF model, HCRTR2-deficient mice exhibited poorer cardiac function, worse treadmill exercise capacity, and greater myocardial scarring. Orexin, an HCRTR2 agonist, rescued function in this HF mouse model. CONCLUSIONS A systems approach identified a novel genetic contribution to human HF and a promising therapeutic agent efficacious in an HF model.


PLOS Genetics | 2016

Discovery of Genetic Variation on Chromosome 5q22 Associated with Mortality in Heart Failure

J. Gustav Smith; Janine F. Felix; Alanna C. Morrison; Andreas P. Kalogeropoulos; Stella Trompet; Jemma B. Wilk; Olof Gidlöf; Xinchen Wang; Michael Morley; Michael M. Mendelson; Roby Joehanes; Symen Ligthart; Xiaoyin Shan; Joshua C. Bis; Ying A. Wang; Marketa Sjögren; Julius S. Ngwa; Jeffrey Brandimarto; David J. Stott; David Aguilar; Kenneth Rice; Howard D. Sesso; Serkalem Demissie; Brendan M. Buckley; Kent D. Taylor; Ian Ford; Chen Yao; Chunyu Liu; Nona Sotoodehnia; Pim van der Harst

Failure of the human heart to maintain sufficient output of blood for the demands of the body, heart failure, is a common condition with high mortality even with modern therapeutic alternatives. To identify molecular determinants of mortality in patients with new-onset heart failure, we performed a meta-analysis of genome-wide association studies and follow-up genotyping in independent populations. We identified and replicated an association for a genetic variant on chromosome 5q22 with 36% increased risk of death in subjects with heart failure (rs9885413, P = 2.7x10-9). We provide evidence from reporter gene assays, computational predictions and epigenomic marks that this polymorphism increases activity of an enhancer region active in multiple human tissues. The polymorphism was further reproducibly associated with a DNA methylation signature in whole blood (P = 4.5x10-40) that also associated with allergic sensitization and expression in blood of the cytokine TSLP (P = 1.1x10-4). Knockdown of the transcription factor predicted to bind the enhancer region (NHLH1) in a human cell line (HEK293) expressing NHLH1 resulted in lower TSLP expression. In addition, we observed evidence of recent positive selection acting on the risk allele in populations of African descent. Our findings provide novel genetic leads to factors that influence mortality in patients with heart failure.


bioRxiv | 2016

A community overlap strategy reveals central genes and networks in heart failure

Pablo Cordero; Ayca Erbilgin; Ching Shang; Michael P. Morley; Matthew T. Wheeler; Frederick E. Dewey; Kevin S. Smith; Ray Hu; Jeffrey Brandimarto; Yichuan Liu; Mingyao Li; Hongzhe Li; Scott Ritter; Sihai Dave Zhao; Komal S. Rathi; Liming Qu; Avinash Das; Stephen B. Montgomery; Sridhar Hannenhalli; Christine S. Moravec; Kenneth B. Margulies; Thomas P. Cappola; Euan A. Ashley

Heart failure is a leading cause of mortality, yet our understanding of the genetic interactions underlying this disease remains incomplete. Here, we harvested 1352 healthy and failing human hearts directly from transplant center operating rooms, and obtained genome-wide genotyping and gene expression measurements for a subset of 313. We built failing and non-failing cardiac regulatory gene networks, revealing important regulators and cardiac expression quantitative trait loci (eQTLs). PPP1R3A emerged as a novel regulator whose network connectivity changed significantly between health and disease. Time-course RNA sequencing after PPP1R3A knock-down validated network-based predictions of metabolic pathway expression, increased cardiomyocyte size, and perturbed respiratory metabolism. Mice lacking PPP1R3A were protected against pressure-overload heart failure. We present a global gene interaction map of the human heart failure transition, identify new cardiac eQTLs, and demonstrate the discovery potential of disease-specific networks through the description of PPP1R3A as a novel central protective regulator in heart failure.Heart failure is one of the leading causes of mortality worldwide, but its underlying molecular mechanisms are poorly understood. To obtain a systems view of the molecular networks that underlie heart failure, we harvested 1352 samples from 313 healthy and failing hearts directly from transplant operating rooms and obtained left-ventricular whole-genome gene expression and genotype measurements. From these data, we built directed regulatory gene networks and gene communities using an approach that combines network and community inference in one framework. Differences in co-expression and global and local centrality parameters pinpointed changes in the molecular interaction network associated with heart failure, as well as its network-wise genetic determinants. Connectivity of one gene, PPP1R3A, previously unassociated with heart failure, changed significantly between healthy and diseased states. Perturbation of in vitro and in vivo systems via time series transcriptome sequencing and murine cardiovascular phenotyping revealed that ablation of PPP1R3A alters disease progression.


JACC: Basic to Translational Science | 2018

Increased Afterload Augments Sunitinib-Induced Cardiotoxicity in an Engineered Cardiac Microtissue Model

Rachel Truitt; Anbin Mu; Elise A. Corbin; Alexia Vite; Jeffrey Brandimarto; Bonnie Ky; Kenneth B. Margulies

Visual Abstract


American Journal of Cardiology | 2016

Effect of Heart Failure With Preserved Ejection Fraction on Nitric Oxide Metabolites.

Payman Zamani; Benjamin French; Jeffrey Brandimarto; Paschalis-Thomas Doulias; Ali Javaheri; Julio A. Chirinos; Kenneth B. Margulies; Raymond R. Townsend; Nancy K. Sweitzer; James C. Fang; Harry Ischiropoulos; Thomas P. Cappola

Collaboration


Dive into the Jeffrey Brandimarto's collaboration.

Top Co-Authors

Avatar

Thomas P. Cappola

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Benjamin French

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bonnie Ky

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Harry Ischiropoulos

Children's Hospital of Philadelphia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Julio A. Chirinos

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge