scispace - formally typeset
Search or ask a question

Showing papers by "David A. Kass published in 2023"


Journal ArticleDOI
TL;DR: In this paper , the authors used agnostic clustering tools, Kruskal-Wallis test with Dunn test, and machine learning to separate heart failure patients with preserved ejection fraction (HFpEF) versus control.
Abstract: Background: The human heart primarily metabolizes fatty acids, and this decreases as alternative fuel use rises in heart failure with reduced ejection fraction (HFrEF). Patients with severe obesity and diabetes are thought to have increased myocardial fatty acid metabolism, but whether this is found in those who also have heart failure with preserved ejection fraction (HFpEF) is unknown. Methods: Plasma and endomyocardial biopsies were obtained from HFpEF (n=38), HFrEF (n=30), and nonfailing donor controls (n=20). Quantitative targeted metabolomics measured organic acids, amino acids, and acylcarnitines in myocardium (72 metabolites) and plasma (69 metabolites). The results were integrated with reported RNA sequencing data. Metabolomics were analyzed using agnostic clustering tools, Kruskal-Wallis test with Dunn test, and machine learning. Results: Agnostic clustering of myocardial but not plasma metabolites separated disease groups. Despite more obesity and diabetes in HFpEF versus HFrEF (body mass index, 39.8 kg/m2 versus 26.1 kg/m2; diabetes, 70% versus 30%; both P<0.0001), medium- and long-chain acylcarnitines (mostly metabolites of fatty acid oxidation) were markedly lower in myocardium from both heart failure groups versus control. In contrast, plasma levels were no different or higher than control. Gene expression linked to fatty acid metabolism was generally lower in HFpEF versus control. Myocardial pyruvate was higher in HFpEF whereas the tricarboxylic acid cycle intermediates succinate and fumarate were lower, as were several genes controlling glucose metabolism. Non–branched-chain and branched-chain amino acids (BCAA) were highest in HFpEF myocardium, yet downstream BCAA metabolites and genes controlling BCAA metabolism were lower. Ketone levels were higher in myocardium and plasma of patients with HFrEF but not HFpEF. HFpEF metabolomic-derived subgroups were differentiated by only a few differences in BCAA metabolites. Conclusions: Despite marked obesity and diabetes, HFpEF myocardium exhibited lower fatty acid metabolites compared with HFrEF. Ketones and metabolites of the tricarboxylic acid cycle and BCAA were also lower in HFpEF, suggesting insufficient use of alternative fuels. These differences were not detectable in plasma and challenge conventional views of myocardial fuel use in HFpEF with marked diabetes and obesity and suggest substantial fuel inflexibility in this syndrome.

5 citations


Journal ArticleDOI
TL;DR: In this article , the authors obtained total proteomic profiles, consisting of 2250 proteins, from right ventricular endomyocardial biopsies in 43 heart failure patients and used machine learning based cluster analysis to identify whether proteomic signatures associate with these deficits.

Journal ArticleDOI
TL;DR: In this paper , the authors performed mechanics analysis on cardiac myofibrils from three common heart failure with preserved ejection fraction (HFpEF) animal models to assess potential differences in myoftibrillar force and relaxation kinetics, independent of higher order deficits associated with the disease.

Journal ArticleDOI
TL;DR: In this article , an unsupervised machine learning approach was used to characterize right ventricular (RV) myocyte contractile depression in patients with heart failure with reduced ejection fraction and pulmonary hypertension (HFrEF-PH), identifying those components reflected by clinical RV indices and uncover underlying biophysical mechanisms.
Abstract: Background: Right ventricular (RV) contractile dysfunction commonly occurs and worsens outcomes in patients with heart failure with reduced ejection fraction and pulmonary hypertension (HFrEF-PH). However, such dysfunction often goes undetected by standard clinical RV indices, raising concerns that they may not reflect aspects of underlying myocyte dysfunction. We thus sought to characterize RV myocyte contractile depression in HFrEF-PH, identify those components reflected by clinical RV indices, and uncover underlying biophysical mechanisms. Methods: Resting, calcium-, and load-dependent mechanics were prospectively studied in permeabilized RV cardiomyocytes isolated from explanted hearts from 23 patients with HFrEF-PH undergoing cardiac transplantation and 9 organ donor controls. Results: Unsupervised machine learning using myocyte mechanical data with the highest variance yielded 2 HFrEF-PH subgroups that in turn mapped to patients with decompensated or compensated clinical RV function. This correspondence was driven by reduced calcium-activated isometric tension in decompensated clinical RV function, whereas surprisingly, many other major myocyte contractile measures including peak power and myocyte active stiffness were similarly depressed in both groups. Similar results were obtained when subgroups were first defined by clinical indices, and then myocyte mechanical properties in each group compared. To test the role of thick filament defects, myofibrillar structure was assessed by x-ray diffraction of muscle fibers. This revealed more myosin heads associated with the thick filament backbone in decompensated clinical RV function, but not compensated clinical RV function, as compared with controls. This corresponded to reduced myosin ATP turnover in decompensated clinical RV function myocytes, indicating less myosin in a crossbridge-ready disordered-relaxed (DRX) state. Altering DRX proportion (%DRX) affected peak calcium-activated tension in the patient groups differently, depending on their basal %DRX, highlighting potential roles for precision-guided therapeutics. Last, increasing myocyte preload (sarcomere length) increased %DRX 1.5-fold in controls but only 1.2-fold in both HFrEF-PH groups, revealing a novel mechanism for reduced myocyte active stiffness and by extension Frank-Starling reserve in human heart failure. Conclusions: Although there are many RV myocyte contractile deficits in HFrEF-PH, commonly used clinical indices only detect reduced isometric calcium-stimulated force, which is related to deficits in basal and recruitable %DRX myosin. Our results support use of therapies to increase %DRX and enhance length-dependent recruitment of DRX myosin heads in such patients.

Proceedings ArticleDOI
18 May 2023
TL;DR: The mtCaMKII interactome was mapped via liquid chromatography-mass spectrometry (LCMS) and proteomics analysis of both scaffolding interactions with proximity labeling utilizing TurboID technology alongside endogenous protein pulldown as discussed by the authors .
Abstract: Abstract ID 29057 Poster Board 155 Ca2+/calmodulin-dependent protein kinase II (CaMKII) is an established negative regulator of cardiac injury. Both the expression and activity levels of CaMKII are elevated in models of heart failure such as ischemia-reperfusion (I/R) injury and myocardial infarction (MI). This is due in part to CaMKII’s role in the regulation of excitation-contraction coupling, apoptosis, activation of hypertrophic programming, arrhythmias and pro-inflammatory signaling. We have recently identified a novel mitochondrial localization for CaMKII (mtCaMKII); importantly, there is an observed increase in mtCaMKII activation with left ventricular dilation following injury in a mouse model of MI. This deleterious post-MI phenotype is rescued with genetic mitochondrial CaMKII inhibition; conversely, mice with myocardial and mitochondrial CaMKII overexpression (mtCaMKII) present with severe dilated cardiomyopathy and decreased ATP production. To date, the molecular mechanisms by which mtCaMKII regulates mitochondrial energetics are not fully elucidated. We have identified changes in the activity of enzymes in the mitochondrial electron transport chain and TCA cycle in response to increased CaMKII levels or activity, indicating a novel and critical role in mitochondrial metabolism for this kinase. We are currently mapping the mtCaMKII interactome via liquid chromatography–mass spectrometry (LCMS) and proteomics analysis of both scaffolding interactions with proximity labeling utilizing TurboID technology alongside endogenous protein pulldowns. Additionally, we are identifying novel CaMKII kinase-substrate relationships using an ATP-analogue labeling, in order to identify metabolic enzymes which may be regulated via post-translational modifications by CaMKII. The identification of previously unknown mitochondrial partners for cardiac CaMKII may uncover promising pharmacological targets for cardiovascular therapeutics, particularly in treating the progression of HF.


Posted ContentDOI
12 Mar 2023-bioRxiv
TL;DR: In this article , the authors used machine learning to identify components of myocyte contractile depression in heart failure patients with reduced ejection fraction and pulmonary hypertension (HFrEF-PH) and elucidate their underlying biophysical mechanisms.
Abstract: Rationale Right ventricular (RV) contractile dysfunction commonly occurs and worsens outcomes in heart failure patients with reduced ejection fraction and pulmonary hypertension (HFrEF-PH). However, such dysfunction often goes undetected by standard clinical RV indices, raising concerns that they may not reflect aspects of underlying myocyte dysfunction. Objective To determine components of myocyte contractile depression in HFrEF-PH, identify those reflected by clinical RV indices, and elucidate their underlying biophysical mechanisms. Methods and Results Resting, calcium- and load-dependent mechanics were measured in permeabilized RV cardiomyocytes isolated from explanted hearts from 23 HFrEF-PH patients undergoing cardiac transplantation and 9 organ-donor controls. Unsupervised machine learning using myocyte mechanical data with the highest variance yielded two HFrEF-PH subgroups that in turn mapped to patients with depressed (RVd) or compensated (RVc) clinical RV function. This correspondence was driven by reduced calcium-activated isometric tension in RVd, while surprisingly, many other major myocyte contractile measures including peak power, maximum unloaded shortening velocity, and myocyte active stiffness were similarly depressed in both groups. Similar results were obtained when subgroups were first defined by clinical indices, and then myocyte mechanical properties in each group compared. To test the role of thick-filament defects, myofibrillar structure was assessed by X-ray diffraction of muscle fibers. This revealed more myosin heads associated with the thick filament backbone in RVd but not RVc, as compared to controls. This corresponded to reduced myosin ATP turnover in RVd myocytes, indicating less myosin in a cross-bridge ready disordered-relaxed (DRX) state. Altering DRX proportion (%DRX) affected peak calcium-activated tension in the patient groups differently, depending on their basal %DRX, highlighting potential roles for precision-guided therapeutics. Lastly, increasing myocyte preload (sarcomere length) increased %DRX 1.5-fold in controls but only 1.2-fold in both HFrEF-PH groups, revealing a novel mechanism for reduced myocyte active stiffness and by extension Frank-Starling reserve in human HF. Conclusions While there are multiple RV myocyte contractile deficits In HFrEF-PH, clinical indices primarily detect reduced isometric calcium-stimulated force related to deficits in basal and recruitable %DRX myosin. Our results support use of therapies to increase %DRX and enhance length-dependent recruitment of DRX myosin heads in such patients.

Journal ArticleDOI
28 Feb 2023-iScience
TL;DR: In this paper , the authors show that O-GlcNAcylated T221 and interactions with coordinating residues are required for normal TRPC6 channel conductance and NFAT activation.