348 results on '"Saucerman, Jeffrey J"'
Search Results
102. Cardiac biexcitability: Two ways to catch a wave
- Author
-
Saucerman, Jeffrey J., primary
- Published
- 2012
- Full Text
- View/download PDF
103. Abstract P203: Nuclear PKA Compartmentation Manages Hypertrophic Responses to β-Adrenergic Signaling
- Author
-
Yang, Jason H, primary and Saucerman, Jeffrey J, additional
- Published
- 2011
- Full Text
- View/download PDF
104. Abstract P206: Automated Imaging Reveals a Switch Between Reversible and Irreversible Cardiac Myocyte Hypertrophy
- Author
-
Ryall, Karen A, primary and Saucerman, Jeffrey J, additional
- Published
- 2011
- Full Text
- View/download PDF
105. Bigger, Better, Faster
- Author
-
Greenwald, Eric C., primary and Saucerman, Jeffrey J., additional
- Published
- 2011
- Full Text
- View/download PDF
106. Graphical Approach to Model Reduction for Nonlinear Biochemical Networks
- Author
-
Holland, David O., primary, Krainak, Nicholas C., additional, and Saucerman, Jeffrey J., additional
- Published
- 2011
- Full Text
- View/download PDF
107. Robustness Portraits of Diverse Biological Networks Conserved Despite Order-Of-Magnitude Parameter Variation
- Author
-
Soltis, Anthony R., primary and Saucerman, Jeffrey J., additional
- Published
- 2011
- Full Text
- View/download PDF
108. Netflux: Biological Network Modeling for Biologists and Students
- Author
-
Dang, Stephen T., primary and Saucerman, Jeffrey J., additional
- Published
- 2011
- Full Text
- View/download PDF
109. Validating a Model of Nitric Oxide-Ca2+ Crosstalk in Cardiac Myocytes
- Author
-
Polanowska-Grabowska, Renata, primary, Ra Park, Sa, additional, and Saucerman, Jeffrey J., additional
- Published
- 2011
- Full Text
- View/download PDF
110. Computational Models Reduce Complexity and Accelerate Insight Into Cardiac Signaling Networks
- Author
-
Yang, Jason H., primary and Saucerman, Jeffrey J., additional
- Published
- 2011
- Full Text
- View/download PDF
111. Cardiac Models in Drug Discovery and Development: A Review
- Author
-
Amanfu, Robert K., primary and Saucerman, Jeffrey J., additional
- Published
- 2011
- Full Text
- View/download PDF
112. Systems Analysis of Small Signaling Modules Relevant to Eight Human Diseases
- Author
-
Benedict, Kelly F., primary, Mac Gabhann, Feilim, additional, Amanfu, Robert K., additional, Chavali, Arvind K., additional, Gianchandani, Erwin P., additional, Glaw, Lydia S., additional, Oberhardt, Matthew A., additional, Thorne, Bryan C., additional, Yang, Jason H., additional, Papin, Jason A., additional, Peirce, Shayn M., additional, Saucerman, Jeffrey J., additional, and Skalak, Thomas C., additional
- Published
- 2010
- Full Text
- View/download PDF
113. Modeling cardiac β-adrenergic signaling with normalized-Hill differential equations: comparison with a biochemical model
- Author
-
Kraeutler, Matthew J, primary, Soltis, Anthony R, additional, and Saucerman, Jeffrey J, additional
- Published
- 2010
- Full Text
- View/download PDF
114. Synergy between CaMKII Substrates and β-Adrenergic Signaling in Regulation of Cardiac Myocyte Ca2+ Handling
- Author
-
Soltis, Anthony R., primary and Saucerman, Jeffrey J., additional
- Published
- 2010
- Full Text
- View/download PDF
115. Modeling Nitric Oxide Regulation Of Ec Coupling In Cardiac Myocytes
- Author
-
Chu, Lulu, primary, Park, Sa Ra, additional, Tandon, Mayank, additional, Guilford, William, additional, and Saucerman, Jeffrey J., additional
- Published
- 2009
- Full Text
- View/download PDF
116. Differential Integration of Ca2+-Calmodulin Signal in Intact Ventricular Myocytes at Low and High Affinity Ca2+-Calmodulin Targets
- Author
-
Song, Qiujing, primary, Saucerman, Jeffrey J., additional, Bossuyt, Julie, additional, and Bers, Donald M., additional
- Published
- 2008
- Full Text
- View/download PDF
117. Calmodulin Mediates Differential Sensitivity of CaMKII and Calcineurin to Local Ca2+ in Cardiac Myocytes
- Author
-
Saucerman, Jeffrey J., primary and Bers, Donald M., additional
- Published
- 2008
- Full Text
- View/download PDF
118. Proarrhythmic Consequences of a KCNQ1 AKAP-Binding Domain Mutation
- Author
-
Saucerman, Jeffrey J., primary, Healy, Sarah N., additional, Belik, Mary E., additional, Puglisi, Jose L., additional, and McCulloch, Andrew D., additional
- Published
- 2004
- Full Text
- View/download PDF
119. Mechanistic systems models of cell signaling networks: a case study of myocyte adrenergic regulation
- Author
-
Saucerman, Jeffrey J., primary and McCulloch, Andrew D., additional
- Published
- 2004
- Full Text
- View/download PDF
120. Modeling β-Adrenergic Control of Cardiac Myocyte Contractility in Silico
- Author
-
Saucerman, Jeffrey J., primary, Brunton, Laurence L., additional, Michailova, Anushka P., additional, and McCulloch, Andrew D., additional
- Published
- 2003
- Full Text
- View/download PDF
121. Mechanisms of cyclic AMP compartmentation revealed by computational models.
- Author
-
Saucerman, Jeffrey J., Greenwald, Eric C., and Polanowska-Grabowska, Renata
- Subjects
- *
CYCLIC adenylic acid , *RADIOIMMUNOASSAY , *ELECTROPHYSIOLOGY , *CELLS , *ADENOSINE monophosphate - Abstract
The article focuses on mechanisms of compartmentation of cyclic adenosine monophosphate (cAMP). The initial measurements of such compartmentation were done via radioimmunoassay and cellular fractionation. The use of patchclamp electrophysiology led to more direct measurement of the compartmentation in living cells.
- Published
- 2014
- Full Text
- View/download PDF
122. Phospholemman is a negative feed-forward regulator of Ca2+ in β-adrenergic signaling, accelerating β-adrenergic inotropy
- Author
-
Yang, Jason H. and Saucerman, Jeffrey J.
- Subjects
- *
PHOSPHOLEMMAN , *CALCIUM ions , *BETA adrenoceptors , *MUSCLE contraction , *CELLULAR signal transduction , *CARDIAC contraction , *PROTEIN kinases , *GENETIC regulation , *SARCOPLASMIC reticulum - Abstract
Abstract: Sympathetic stimulation enhances cardiac contractility by stimulating β-adrenergic signaling and protein kinase A (PKA). Recently, phospholemman (PLM) has emerged as an important PKA substrate capable of regulating cytosolic Ca2+ transients. However, it remains unclear how PLM contributes to β-adrenergic inotropy. Here we developed a computational model to clarify PLM''s role in the β-adrenergic signaling response. Simulating Na+ and sarcoplasmic reticulum (SR) Ca2+ clamps, we identify an effect of PLM phosphorylation on SR unloading as the key mechanism by which PLM confers cytosolic Ca2+ adaptation to long-term β-adrenergic receptor (β-AR) stimulation. Moreover, we show that phospholamban (PLB) opposes and overtakes these actions on SR load, forming a negative feed-forward loop in the β-adrenergic signaling cascade. This network motif dominates the negative feedback conferred by β-AR desensitization and accelerates β-AR-induced inotropy. Model analysis therefore unmasks key actions of PLM phosphorylation during β-adrenergic signaling, indicating that PLM is a critical component of the fight-or-flight response. [Copyright &y& Elsevier]
- Published
- 2012
- Full Text
- View/download PDF
123. Calmodulin binding proteins provide domains of local Ca2+ signaling in cardiac myocytes
- Author
-
Saucerman, Jeffrey J. and Bers, Donald M.
- Subjects
- *
CALMODULIN , *MUSCLE cells , *CALCIUM channels , *CARRIER proteins , *CELLULAR signal transduction , *HEALTH outcome assessment - Abstract
Abstract: Calmodulin (CaM) acts as a common Ca2+ sensor for many signaling pathways, transducing local Ca2+ signals into specific cellular outcomes. Many of CaM''s signaling functions can be explained by its unique biochemical properties, including high and low affinity Ca2+-binding sites with slow and fast kinetics, respectively. CaM is expected to have a limited spatial range of action, emphasizing its role in local Ca2+ signaling. Interactions with target proteins further fine-tune CaM signal transduction. Here, we focus on only three specific cellular targets for CaM signaling in cardiac myocytes: the L-type Ca2+ channel, the ryanodine receptor, and the IP3 receptor. We elaborate a working hypothesis that each channel is regulated by two distinct functional populations of CaM: dedicated CaM and promiscuous CaM. Dedicated CaM is typically tethered to each channel and directly regulates channel activity. In addition, a local pool of promiscuous CaM appears poised to sense local Ca2+ signals and trigger downstream pathways such as Ca2+/CaM dependent-protein kinase II and calcineurin. Understanding how promiscuous CaM coordinates multiple distinct signaling pathways remains a challenge, but is aided by the use of mathematical modeling and a new generation of fluorescent biosensors. This article is part of a special issue entitled "Local Signaling in Myocytes." [Copyright &y& Elsevier]
- Published
- 2012
- Full Text
- View/download PDF
124. Modeling cardiac β-adrenergic signaling withnormalized-Hill differential equations: comparisonwith a biochemical model.
- Author
-
Kraeutler, Matthew J., Soltis, Anthony R., and Saucerman, Jeffrey J.
- Subjects
CELLULAR signal transduction ,DIFFERENTIAL equations ,BIOLOGICAL models ,PHOSPHODIESTERASES ,PHYSIOLOGICAL control systems - Abstract
Background: New approaches are needed for large-scale predictive modeling of cellular signaling networks. While mass action and enzyme kinetic approaches require extensive biochemical data, current logic-based approaches are used primarily for qualitative predictions and have lacked direct quantitative comparison with biochemical models. Results: We developed a logic-based differential equation modeling approach for cell signaling networks based on normalized Hill activation/inhibition functions controlled by logical AND and OR operators to characterize signaling crosstalk. Using this approach, we modeled the cardiac β
1 -adrenergic signaling network, including 36 reactions and 25 species. Direct comparison of this model to an extensively characterized and validated biochemical model of the same network revealed that the new model gave reasonably accurate predictions of key network properties, even with default parameters. Normalized Hill functions improved quantitative predictions of global functional relationships compared with prior logic-based approaches. Comprehensive sensitivity analysis revealed the significant role of PKA negative feedback on upstream signaling and the importance of phosphodiesterases as key negative regulators of the network. The model was then extended to incorporate recently identified protein interaction data involving integrin-mediated mechanotransduction. Conclusions: The normalized-Hill differential equation modeling approach allows quantitative prediction of network functional relationships and dynamics, even in systems with limited biochemical data. [ABSTRACT FROM AUTHOR]- Published
- 2010
- Full Text
- View/download PDF
125. Multiscale Modeling in Rodent Ventricular Myocytes.
- Author
-
Shaoying Lu, Michailova, Anushka P., Saucerman, Jeffrey J., Yuhui Cheng, Zeyun Yu, Kaiser, Timothy H., Li, Wilfred W., Bank, Randolph E., Holst, Michael J., McCammon, J. Andrew, Hayashi, Takeharu, Hoshijima, Masahiko, Arzberger, Peter, and McCulloch, Andrew D.
- Subjects
MUSCLE cells ,CALCIUM ions ,LABORATORY rats ,DIFFUSION processes ,VENTRICULAR remodeling ,MYOCARDIAL infarction complications ,EXCITABLE membranes ,HETEROGENEITY ,EXCITATION (Physiology) - Abstract
The article presents a discussion on the developed three dimensional (3-D) continuum model used for the laboratory observation of calcium ion (Ca
2+ ) properties on rat myocytes. It provides information on the geometrical model and structural data observed and gathered on the rat's ventricular muscle cell. It also investigates mechanisms involving the basic principles of excitation-contraction (EC) coupling propagation in the observed ventricular myocytes. The study also reveals that the local Ca2+ spatiotemporal features signals that relies on the axial and cell surface diffusion distances.- Published
- 2009
- Full Text
- View/download PDF
126. Differential Integration of Ca2+-Calmodulin Signal in Intact Ventricular Myocytes at Low and High Affinity Ca2+-Calmodulin Targets.
- Author
-
Qiujing Song, Saucerman, Jeffrey J., Bossuyt, Julie, and Bers, Donald M.
- Subjects
- *
MUSCLE cells , *CALMODULIN , *CALCIUM-binding proteins , *PROTEIN kinases , *BIOSENSORS , *BIOCHEMISTRY - Abstract
Cardiac myocyte intracellular calcium varies beat-to-beat and calmodulin (CaM) transduces Ca2+ signals to regulate many cellular processes (e.g. via CaM targets such as CaM-dependent kinase and calcineurin). However, little is known about the dynamics of how CaM targets process the Ca2+ signals to generate appropriate biological responses in the heart. We hypothesized that the different affinities of CaM targets for the Ca2+-bound CaM (Ca2+-CaM) shape their actions through dynamic and tonic interactions in response to the repetitive Ca2+ signals in myocytes. To test our hypothesis, we used two fluorescence resonance energy transfer-based biosensors, BsCaM-45 (Kd = ∼45 nM) and BsCaM-2 (Kd = ∼2 nM), to monitor the real time Ca2+-CaM dynamics at low and high affinity CaM targets in paced adult ventricular myocytes. Compared with BsCaM-2, BsCaM-45 tracks the beat-to-beat Ca2+-CaM alterations more closely following the Ca2+ oscillations at each myocyte contraction. When pacing frequency is raised from 0.1 to 1.0 Hz, the higher affinity BsCaM-2 demonstrates significant elevation of diastolic Ca2+-CaM binding compared with the lower affinity BsCaM-45. Biochemically detailed computational models of Ca2+-CaM biosensors in beating cardiac myocytes revealed that the different Ca2+-CaM binding affinities of BsCaM-2 and BsCaM-45 are sufficient to predict their differing kinetics and diastolic integration. Thus, data from both experiments and computational modeling suggest that CaM targets with low versus high Ca2+-CaM affinities (like CaM-dependent kinase versus calcineurin) respond differentially to the same Ca2+ signal (phasic versus integrating), presumably tuned appropriately for their respective and distinct Ca2+ signaling pathways. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
127. Identification and Characterization of Poly(I:C)-induced Molecular Responses Attenuated by Nicotine in Mouse Macrophages
- Author
-
Cui, Wen-Yan, Zhao, Shufang, Polanowska-Grabowska, Renata, Wang, Ju, Wei, Jinxue, Dash, Bhagirathi, Chang, Sulie L., Saucerman, Jeffrey J., Gu, Jun, and Li, Ming D.
- Abstract
To further our understanding of the effects of nicotine on the molecular responses of macrophages during virus or virus-like infections, poly(I:C)-stimulated macrophage-like RAW264.2 cells or mouse primary peritoneal macrophages were challenged with nicotine; and their molecular responses were evaluated using a qRT-PCR array, antibody array, ELISA, Western blotting, and Ca2+imaging. Of 51 genes expressed in the Toll-like receptor (TLR) and RIG-I-like receptor (RLR) pathways, mRNA expression of 15 genes in RAW264.7 cells was attenuated by nicotine, of which mRNA expression of IL-6, TNF-α, and IL-1βwas confirmed to be attenuated in peritoneal macrophages. Concurrently, nicotine treatment attenuated the release of IL-6 and TNF-αfrom poly(I:C)-stimulated macrophages. However, when poly(I:C)-stimulated macrophages were challenged with nicotine plus α-bungarotoxin (α-BTX), secretion of IL-6 and TNF-αwas found to be in a level seen with poly(I:C) stimulation only, indicating that α7-nAChR, a highly Ca2+permeable ion channel sensitive to blockade by α-BTX, is involved in this process. Furthermore, results from an antibody array indicated that nicotine treatment attenuated the phosphorylation of 82 sites, including Thr286 on CaMKIIα, from poly(I:C)-stimulated RAW264.7 cells, of which 28 are expressed in the downstream cascade of Ca2+signaling. Coincidentally, poly(I:C)-stimulated macrophages showed attenuated expression of phosphorylated CaMKIIαwhen pretreated with nicotine. In addition, nicotine attenuated intracellular Ca2+signal from poly(I:C)-stimulated RAW264.7 cells. Collectively, these results indicate that poly(I:C)-induced molecular responses of macrophages could be significantly attenuated by nicotine.
- Published
- 2013
- Full Text
- View/download PDF
128. Mechano-chemo signaling interactions modulate matrix production by cardiac fibroblasts
- Author
-
Rogers, Jesse D., Holmes, Jeffrey W., Saucerman, Jeffrey J., and Richardson, William J.
- Abstract
Extracellular matrix remodeling after myocardial infarction occurs in a dynamic environment in which local mechanical stresses and biochemical signaling species stimulate the accumulation of collagen-rich scar tissue. It is well-known that cardiac fibroblasts regulate post-infarction matrix turnover by secreting matrix proteins, proteases, and protease inhibitors in response to both biochemical stimuli and mechanical stretch, but how these stimuli act together to dictate cellular responses is still unclear. We developed a screen of cardiac fibroblast-secreted proteins in response to combinations of biochemical agonists and cyclic uniaxial stretch in order to elucidate the relationships between stretch, biochemical signaling, and cardiac matrix turnover. We found that stretch significantly synergized with biochemical agonists to inhibit the secretion of matrix metalloproteinases, with stretch either amplifying protease suppression by individual agonists or antagonizing agonist-driven upregulation of protease expression. Stretch also modulated fibroblast sensitivity towards biochemical agonists by either sensitizing cells towards agonists that suppress protease secretion or de-sensitizing cells towards agonists that upregulate protease secretion. These findings suggest that the mechanical environment can significantly alter fibrosis-related signaling in cardiac fibroblasts, suggesting caution when extrapolating in vitro data to predict effects of fibrosis-related cytokines in situations like myocardial infarction where mechanical stretch occurs.
- Published
- 2021
- Full Text
- View/download PDF
129. A personalized, multiomics approach identifies genes involved in cardiac hypertrophy and heart failure.
- Author
-
Santolini, Marc, Romay, Milagros C., Yukhtman, Clara L., Rau, Christoph D., Ren, Shuxun, Saucerman, Jeffrey J., Wang, Jessica J., Weiss, James N., Wang, Yibin, Lusis, Aldons J., and Karma, Alain
- Subjects
CARDIAC hypertrophy ,HEART failure ,GENE expression ,PHENOTYPES ,INDIVIDUALIZED medicine - Abstract
A traditional approach to investigate the genetic basis of complex diseases is to identify genes with a global change in expression between diseased and healthy individuals. However, population heterogeneity may undermine the effort to uncover genes with significant but individual contribution to the spectrum of disease phenotypes within a population. Here we investigate individual changes of gene expression when inducing hypertrophy and heart failure in 100 + strains of genetically distinct mice from the Hybrid Mouse Diversity Panel (HMDP). We find that genes whose expression fold-change correlates in a statistically significant way with the severity of the disease are either up or down-regulated across strains, and therefore missed by a traditional population-wide analysis of differential gene expression. Furthermore, those "fold-change" genes are enriched in human cardiac disease genes and form a dense co-regulated module strongly interacting with the cardiac hypertrophic signaling network in the human interactome. We validate our approach by showing that the knockdown of Hes1, predicted as a strong candidate, induces a dramatic reduction of hypertrophy by 80–90% in neonatal rat ventricular myocytes. Our results demonstrate that individualized approaches are crucial to identify genes underlying complex diseases as well as to develop personalized therapies. Personalized medicine: uncovering missed disease genes A multitude of genes associated with complex diseases are revealed by a novel personalized, as opposed to population-level, analysis of differential gene expression. While traditional investigations of the genetic basis of complex diseases assume homogeneity across individuals and identify genes differentially expressed between a diseased and a healthy population, Northeastern University and University of California Los Angeles researchers have identified a different class of disease genes that exhibit heterogeneous up and down-regulation across 100 genetically distinct mouse strains subject to a stressor inducing heart failure, but show no significant change of expression at the population level. The results, validated by in vitro knockdown, demonstrate that individualized approaches are crucial to unmask all genes involved in complex diseases, opening new avenues for the development of personalized therapies. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
130. A multiscale model of cardiac concentric hypertrophy incorporating both mechanical and hormonal drivers of growth.
- Author
-
Estrada, Ana C., Yoshida, Kyoko, Saucerman, Jeffrey J., and Holmes, Jeffrey W.
- Subjects
- *
MULTISCALE modeling , *CARDIAC hypertrophy , *ANGIOTENSIN receptors , *GENETIC engineering , *PROPRANOLOL , *POWER transmission - Abstract
Growth and remodeling in the heart is driven by a combination of mechanical and hormonal signals that produce different patterns of growth in response to exercise, pregnancy, and various pathologies. In particular, increases in afterload lead to concentric hypertrophy, a thickening of the walls that increases the contractile ability of the heart while reducing wall stress. In the current study, we constructed a multiscale model of cardiac hypertrophy that connects a finite-element model representing the mechanics of the growing left ventricle to a cell-level network model of hypertrophic signaling pathways that accounts for changes in both mechanics and hormones. We first tuned our model to capture published in vivo growth trends for isoproterenol infusion, which stimulates β-adrenergic signaling pathways without altering mechanics, and for transverse aortic constriction (TAC), which involves both elevated mechanics and altered hormone levels. We then predicted the attenuation of TAC-induced hypertrophy by two distinct genetic interventions (transgenic Gq-coupled receptor inhibitor overexpression and norepinephrine knock-out) and by two pharmacologic interventions (angiotensin receptor blocker losartan and β-blocker propranolol) and compared our predictions to published in vivo data for each intervention. Our multiscale model captured the experimental data trends reasonably well for all conditions simulated. We also found that when prescribing realistic changes in mechanics and hormones associated with TAC, the hormonal inputs were responsible for the majority of the growth predicted by the multiscale model and were necessary in order to capture the effect of the interventions for TAC. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
131. Identifying metabolic adaptations characteristic of cardiotoxicity using paired transcriptomics and metabolomics data integrated with a computational model of heart metabolism.
- Author
-
Dougherty, Bonnie V., Moore, Connor J., Rawls, Kristopher D., Jenior, Matthew L., Chun, Bryan, Nagdas, Sarbajeet, Saucerman, Jeffrey J., Kolling, Glynis L., Wallqvist, Anders, and Papin, Jason A.
- Subjects
- *
HEART metabolism , *CARDIOTOXICITY , *TRANSCRIPTOMES , *METABOLIC models , *METABOLOMICS , *RNA metabolism - Abstract
Improvements in the diagnosis and treatment of cancer have revealed long-term side effects of chemotherapeutics, particularly cardiotoxicity. Here, we present paired transcriptomics and metabolomics data characterizing in vitro cardiotoxicity to three compounds: 5-fluorouracil, acetaminophen, and doxorubicin. Standard gene enrichment and metabolomics approaches identify some commonly affected pathways and metabolites but are not able to readily identify metabolic adaptations in response to cardiotoxicity. The paired data was integrated with a genome-scale metabolic network reconstruction of the heart to identify shifted metabolic functions, unique metabolic reactions, and changes in flux in metabolic reactions in response to these compounds. Using this approach, we confirm previously seen changes in the p53 pathway by doxorubicin and RNA synthesis by 5-fluorouracil, we find evidence for an increase in phospholipid metabolism in response to acetaminophen, and we see a shift in central carbon metabolism suggesting an increase in metabolic demand after treatment with doxorubicin and 5-fluorouracil. Author summary: Improvements in the diagnosis and treatment of cancer have revealed long-term side effects of chemotherapeutics, particularly cardiotoxicity. Here, we compare the cardiotoxic effects of 3 drugs, 5-fluorouracil, acetaminophen, and doxorubicin, using transcriptomic and metabolomic data. By integrating these data into a genome-scale metabolic network reconstruction of the heart, we were able to identify metabolic adaptations in response to cardiotoxicity that avoided detection in traditional gene enrichment and metabolomic techniques. Using this approach, we confirm known mechanisms of doxorubicin-induced cardiotoxicity and provide hypotheses and potential mechanisms for metabolic adaptations in cardiotoxicity for 5-fluorouracil, doxorubicin, and acetaminophen. We hope future work using our novel paired transcriptomic and metabolic characterization of in vitro cardiotoxicity can help improve the current network heart model and further characterize the role of chemotherapeutics in cardiotoxicity. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
132. Differential sensitivity to longitudinal and transverse stretch mediates transcriptional responses in mouse neonatal ventricular myocytes.
- Author
-
Shulin Cao, Buchholz, Kyle S., Tan, Philip, Stowe, Jennifer C., Wang, Ariel, Fowler, Annabelle, Knaus, Katherine R., Khalilimeybodi, Ali, Zambon, Alexander C., Omens, Jeffrey H., Saucerman, Jeffrey J., and McCulloch, Andrew D.
- Subjects
- *
GENETIC regulation , *CALCIUM channels , *GENE expression , *MITOGEN-activated protein kinases , *GENETIC transcription regulation - Abstract
To identify how cardiomyocyte mechanosensitive signaling pathways are regulated by anisotropic stretch, micropatterned mouse neonatal cardiomyocytes were stretched primarily longitudinally or transversely to the myofiber axis. Four hours of static, longitudinal stretch induced differential expression of 557 genes, compared with 30 induced by transverse stretch, measured using RNA-seq. A logic-based ordinary differential equation model of the cardiac myocyte mechanosignaling network, extended to include the transcriptional regulation and expression of 784 genes, correctly predicted measured expression changes due to anisotropic stretch with 69% accuracy. The model also predicted published transcriptional responses to mechanical load in vitro or in vivo with 63-91% accuracy. The observed differences between transverse and longitudinal stretch responses were not explained by differential activation of specific pathways but rather by an approximately twofold greater sensitivity to longitudinal stretch than transverse stretch. In vitro experiments confirmed model predictions that stretch-induced gene expression is more sensitive to angiotensin II and endothelin-1, via RhoA and MAP kinases, than to the three membrane ion channels upstream of calcium signaling in the network. Quantitative cardiomyocyte gene expression differs substantially with the axis of maximum principal stretch relative to the myofilament axis, but this difference is due primarily to differences in stretch sensitivity rather than to selective activation of mechanosignaling pathways. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
133. Logic-based mechanistic machine learning on high-content images reveals how drugs differentially regulate cardiac fibroblasts.
- Author
-
Nelson, Anders R., Christiansen, Steven L., Naegle, Kristen M., and Saucerman, Jeffrey J.
- Subjects
- *
MACHINE learning , *FIBROBLASTS , *EXTRACELLULAR matrix , *HEART fibrosis , *HEART injuries - Abstract
Fibroblasts are essential regulators of extracellular matrix deposition following cardiac injury. These cells exhibit highly plastic responses in phenotype during fibrosis in response to environmental stimuli. Here, we test whether and how candidate anti-fibrotic drugs differentially regulate measures of cardiac fibroblast phenotype, which may help identify treatments for cardiac fibrosis. We conducted a high-content microscopy screen of human cardiac fibroblasts treated with 13 clinically relevant drugs in the context of TGFß and/or IL-1ß, measuring phenotype across 137 single-cell features. We used the phenotypic data from our high-content imaging to train a logic-based mechanistic machine learning model (LogiMML) for fibroblast signaling. The model predicted how pirfenidone and Src inhibitor WH-4- 023 reduce actin filament assembly and actin-myosin stress fiber formation, respectively. Validating the LogiMML model prediction that PI3K partially mediates the effects of Src inhibition, we found that PI3K inhibition reduces actin-myosin stress fiber formation and procollagen I production in human cardiac fibroblasts. In this study, we establish a modeling approach combining the strengths of logic-based network models and regularized regression models. We apply this approach to predict mechanisms that mediate the differential effects of drugs on fibroblasts, revealing Src inhibition acting via PI3K as a potential therapy for cardiac fibrosis. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
134. THE EFFECT OF MIDSOLE PLUGS INSERTED INTO THERAPEUTIC FOOTWEAR FOR LOCALIZED PLANTAR PRESSURE RELIEF.
- Author
-
Saucerman, Jeffrey J., Loppnow, Brian W., Lemmon, David R., Smoluk, John R., and Cavanagh, Peter R.
- Published
- 2000
135. Network-based predictions of in vivo cardiac hypertrophy.
- Author
-
Frank, Deborah U., Sutcliffe, Matthew D., and Saucerman, Jeffrey J.
- Subjects
- *
CARDIAC hypertrophy , *MYOCARDIUM , *HEART failure , *CELL culture , *PHENOTYPES , *TRANSGENIC mice - Abstract
Cardiac hypertrophy is a common response of cardiac myocytes to stress and a predictor of heart failure. While in vitro cell culture studies have identified numerous molecular mechanisms driving hypertrophy, it is unclear to what extent these mechanisms can be integrated into a consistent framework predictive of in vivo phenotypes. To address this question, we investigate the degree to which an in vitro- based, manually curated computational model of the hypertrophy signaling network is able to predict in vivo hypertrophy of 52 cardiac-specific transgenic mice. After minor revisions motivated by in vivo literature, the model concordantly predicts the qualitative responses of 78% of output species and 69% of signaling intermediates within the network model. Analysis of four double-transgenic mouse models reveals that the computational model robustly predicts hypertrophic responses in mice subjected to multiple, simultaneous perturbations. Thus the model provides a framework with which to mechanistically integrate data from multiple laboratories and experimental systems to predict molecular regulation of cardiac hypertrophy. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
136. Regulation of nuclear PKA revealed by spatiotemporal manipulation of cyclic AMP.
- Author
-
Sample, Vedangi, DiPilato, Lisa M, Yang, Jason H, Ni, Qiang, Saucerman, Jeffrey J, and Zhang, Jin
- Subjects
SPATIOTEMPORAL processes ,CYCLIC adenylic acid - Abstract
A correction to the article "Regulation of nuclear PKA revealed by spatiotemporal manipulation of cyclic AMP" that was published online on February 26, 2012 is presented.
- Published
- 2013
- Full Text
- View/download PDF
137. Signaling network model of cardiomyocyte morphological changes in familial cardiomyopathy.
- Author
-
Khalilimeybodi, Ali, Riaz, Muhammad, Campbell, Stuart G., Omens, Jeffrey H., McCulloch, Andrew D., Qyang, Yibing, and Saucerman, Jeffrey J.
- Subjects
- *
CARDIOMYOPATHIES , *CARDIAC arrest , *CONNECTIN , *GROWTH factors , *CYTOSKELETAL proteins , *CARDIOVASCULAR agents , *GENETIC mutation - Abstract
Familial cardiomyopathy is a precursor of heart failure and sudden cardiac death. Over the past several decades, researchers have discovered numerous gene mutations primarily in sarcomeric and cytoskeletal proteins causing two different disease phenotypes: hypertrophic (HCM) and dilated (DCM) cardiomyopathies. However, molecular mechanisms linking genotype to phenotype remain unclear. Here, we employ a systems approach by integrating experimental findings from preclinical studies (e.g. , murine data) into a cohesive signaling network to scrutinize genotype to phenotype mechanisms. We developed an HCM/DCM signaling network model utilizing a logic-based differential equations approach and evaluated model performance in predicting experimental data from four contexts (HCM, DCM, pressure overload, and volume overload). The model has an overall prediction accuracy of 83.8%, with higher accuracy in the HCM context (90%) than DCM (75%). Global sensitivity analysis identifies key signaling reactions, with calcium-mediated myofilament force development and calcium-calmodulin kinase signaling ranking the highest. A structural revision analysis indicates potential missing interactions that primarily control calcium regulatory proteins, increasing model prediction accuracy. Combination pharmacotherapy analysis suggests that downregulation of signaling components such as calcium, titin and its associated proteins, growth factor receptors, ERK1/2, and PI3K-AKT could inhibit myocyte growth in HCM. In experiments with patient-specific iPSC-derived cardiomyocytes (MLP-W4R;MYH7-R723C iPSC-CMs), combined inhibition of ERK1/2 and PI3K-AKT rescued the HCM phenotype, as predicted by the model. In DCM, PI3K-AKT-NFAT downregulation combined with upregulation of Ras/ERK1/2 or titin or Gq protein could ameliorate cardiomyocyte morphology. The model results suggest that HCM mutations that increase active force through elevated calcium sensitivity could increase ERK activity and decrease eccentricity through parallel growth factors, Gq-mediated, and titin pathways. Moreover, the model simulated the influence of existing medications on cardiac growth in HCM and DCM contexts. This HCM/DCM signaling model demonstrates utility in investigating genotype to phenotype mechanisms in familial cardiomyopathy. [Display omitted] • HCM/DCM signaling network model links mutations to cardiomyocyte morphological changes. • The computational model predicts higher efficiency for combination pharmacotherapies in familial cardiomyopathy. • Experiments on iPSC-CMs showed rescue of HCM phenotype by combined inhibition of ERK1/2 and PI3K-AKT predicted by the model. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
138. Validating a Model of Nitric Oxide-Ca2+Crosstalk in Cardiac Myocytes
- Author
-
Polanowska-Grabowska, Renata, Ra Park, Sa, and Saucerman, Jeffrey J.
- Published
- 2011
- Full Text
- View/download PDF
139. Computational model of cardiomyocyte apoptosis identifies mechanisms of tyrosine kinase inhibitor-induced cardiotoxicity.
- Author
-
Grabowska, Monika E., Chun, Bryan, Moya, Raquel, and Saucerman, Jeffrey J.
- Subjects
- *
PROTEIN-tyrosine kinases , *MYOCARDIAL reperfusion , *PROTEIN-tyrosine kinase inhibitors , *CARDIOTOXICITY , *APOPTOSIS , *REACTIVE oxygen species - Abstract
Despite clinical observations of cardiotoxicity among cancer patients treated with tyrosine kinase inhibitors (TKIs), the molecular mechanisms by which these drugs affect the heart remain largely unknown. Mechanistic understanding of TKI-induced cardiotoxicity has been limited in part due to the complexity of tyrosine kinase signaling pathways and the multi-targeted nature of many of these drugs. TKI treatment has been associated with reactive oxygen species generation, mitochondrial dysfunction, and apoptosis in cardiomyocytes. To gain insight into the mechanisms mediating TKI-induced cardiotoxicity, this study constructs and validates a computational model of cardiomyocyte apoptosis, integrating intrinsic apoptotic and tyrosine kinase signaling pathways. The model predicts high levels of apoptosis in response to sorafenib, sunitinib, ponatinib, trastuzumab, and gefitinib, and lower levels of apoptosis in response to nilotinib and erlotinib, with the highest level of apoptosis induced by sorafenib. Knockdown simulations identified AP1, ASK1, JNK, MEK47, p53, and ROS as positive functional regulators of sorafenib-induced apoptosis of cardiomyocytes. Overexpression simulations identified Akt, IGF1, PDK1, and PI3K among the negative functional regulators of sorafenib-induced cardiomyocyte apoptosis. A combinatorial screen of the positive and negative regulators of sorafenib-induced apoptosis revealed ROS knockdown coupled with overexpression of FLT3, FGFR, PDGFR, VEGFR, or KIT as a particularly potent combination in reducing sorafenib-induced apoptosis. Network simulations of combinatorial treatment with sorafenib and the antioxidant N -acetyl cysteine (NAC) suggest that NAC may protect cardiomyocytes from sorafenib-induced apoptosis. • Computational model of cardiomyocyte apoptosis signaling network. • The model is validated against a wide range of experimental data. • Virtual screen predicts regulators of tyrosine kinase inhibitor-induced apoptosis. • Inhibition of ROS is predicted to block apoptosis from tyrosine kinase inhibitors. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
140. Network Analysis Reveals a Distinct Axis of Macrophage Activation in Response to Conflicting Inflammatory Cues.
- Author
-
Xiaji Liu, Jingyuan Zhang, Zeigler, Angela C., Nelson, Anders R., Lindsey, Merry L., and Saucerman, Jeffrey J.
- Subjects
- *
MACROPHAGE activation , *INFLAMMATION , *STAT proteins , *CELLULAR signal transduction , *MACROPHAGES - Abstract
Macrophages are subject to a wide range of cytokine and pathogen signals in vivo, which contribute to differential activation and modulation of inflammation. Understanding the response to multiple, often-conflicting cues that macrophages experience requires a network perspective. In this study, we integrate data from literature curation andmRNA expression profiles obtained from wild type C57/BL6J mice macrophages to develop a large-scale computational model of the macrophage signaling network. In response to stimulation across all pairs of nine cytokine inputs, the model predicted activation along the classic M1–M2 polarization axis but also a second axis of macrophage activation that distinguishes unstimulated macrophages from a mixed phenotype induced by conflicting cues. Along this second axis, combinations of conflicting stimuli, IL-4 with LPS, IFN-g, IFN-b, or TNF-a, produced mutual inhibition of several signaling pathways, e.g., NF-kB and STAT6, but also mutual activation of the PI3K signaling module. In response to combined IFN-g and IL-4, the model predicted genes whose expression was mutually inhibited, e.g., iNOS or Nos2 and Arg1, or mutually enhanced, e.g., Il4ra and Socs1, validated by independent experimental data. Knockdown simulations further predicted network mechanisms underlying functional cross-talk, such as mutual STAT3/STAT6-mediated enhancement of Il4ra expression. In summary, the computational model predicts that network cross-talk mediates a broadened spectrum of macrophage activation in response to mixed pro- and anti-inflammatory cytokine cues, making it useful for modeling in vivo scenarios. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
141. Quantification of model and data uncertainty in a network analysis of cardiac myocyte mechanosignalling.
- Author
-
Cao, Shulin, Aboelkassem, Yasser, Wang, Ariel, Valdez-Jasso, Daniela, Saucerman, Jeffrey J., Omens, Jeffrey H., and McCulloch, Andrew D.
- Subjects
- *
MONTE Carlo method , *ORDINARY differential equations , *POLYNOMIAL chaos , *UNCERTAINTY , *EPISTEMIC uncertainty , *DATA modeling - Abstract
Cardiac myocytes transduce changes in mechanical loading into cellular responses via interacting cell signalling pathways. We previously reported a logicbased ordinary differential equation model of the myocyte mechanosignalling network that correctly predicts 78% of independent experimental results not used to formulate the original model. Here, we use Monte Carlo and polynomial chaos expansion simulations to examine the effects of uncertainty in parameter values, model logic and experimental validation data on the assessed accuracy of that model. The prediction accuracy of the model was robust to parameter changes over a wide range being least sensitive to uncertainty in time constants and most affected by uncertainty in reaction weights. Quantifying epistemic uncertainty in the reaction logic of the model showed that while replacing 'OR' with 'AND' reactions greatly reduced model accuracy, replacing 'AND' with 'OR' reactions was more likely to maintain or even improve accuracy. Finally, data uncertainty had a modest effect on assessment of model accuracy. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
142. High-content phenotypic assay for proliferation of human iPSC-derived cardiomyocytes identifies L-type calcium channels as targets.
- Author
-
Woo, Laura A., Tkachenko, Svyatoslav, Ding, Mei, Plowright, Alleyn T., Engkvist, Ola, Andersson, Henrik, Drowley, Lauren, Barrett, Ian, Firth, Mike, Akerblad, Peter, Wolf, Matthew J., Bekiranov, Stefan, Brautigan, David L., Wang, Qing-Dong, and Saucerman, Jeffrey J.
- Subjects
- *
INDUCED pluripotent stem cells , *CELL proliferation , *HEART cells , *CALCIUM channels , *HEART failure , *PUBLIC health - Abstract
Abstract Over 5 million people in the United States suffer from heart failure, due to the limited ability to regenerate functional cardiac tissue. One potential therapeutic strategy is to enhance proliferation of resident cardiomyocytes. However, phenotypic screening for therapeutic agents is challenged by the limited ability of conventional markers to discriminate between cardiomyocyte proliferation and endoreplication (e.g. polyploidy and multinucleation). Here, we developed a novel assay that combines automated live-cell microscopy and image processing algorithms to discriminate between proliferation and endoreplication by quantifying changes in the number of nuclei, changes in the number of cells, binucleation, and nuclear DNA content. We applied this assay to further prioritize hits from a primary screen for DNA synthesis, identifying 30 compounds that enhance proliferation of human induced pluripotent stem cell-derived cardiomyocytes. Among the most active compounds from the phenotypic screen are clinically approved L-type calcium channel blockers from multiple chemical classes whose activities were confirmed across different sources of human induced pluripotent stem cell-derived cardiomyocytes. Identification of compounds that stimulate human cardiomyocyte proliferation may provide new therapeutic strategies for heart failure. Graphical abstract Unlabelled Image Highlights • Novel automated hybrid live/fixed assay for cardiomyocyte proliferation. • Assay distinguishes proliferation from multinucleation and polyploidization. • Assay applied for high-content screening of compounds. • L-type calcium channel blockers induce cardiomyocyte proliferation. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
143. Atg2, Atg9 and Atg18 in mitochondrial integrity, cardiac function and healthspan in Drosophila.
- Author
-
Xu, Peng, Damschroder, Deena, Zhang, Mei, Ryall, Karen A., Adler, Paul N., Saucerman, Jeffrey J., Wessells, Robert J., and Yan, Zhen
- Subjects
- *
MITOCHONDRIA , *EUKARYOTES , *RNA interference , *DROSOPHILA melanogaster , *CARDIAC hypertrophy - Abstract
Abstract In yeast, the Atg2-Atg18 complex regulates Atg9 recycling from phagophore assembly site during autophagy; their function in higher eukaryotes remains largely unknown. In a targeted screening in Drosophila melanogaster , we show that Mef2-GAL4 -RNAi-mediated knockdown of Atg2, Atg9 or Atg18 in the heart and indirect flight muscles led to shortened healthspan (declined locomotive function) and lifespan. These flies displayed an accelerated age-dependent loss of cardiac function along with cardiac hypertrophy (increased heart tube wall thickness) and structural abnormality (distortion of the lumen surface). Using the Mef2-GAL4-MitoTimer mitochondrial reporter system and transmission electron microscopy, we observed significant elongation of mitochondria and reduced number of lysosome-targeted autophagosomes containing mitochondria in the heart tube but exaggerated mitochondrial fragmentation and reduced mitochondrial density in indirect flight muscles. These findings provide the first direct evidence of the importance of Atg2-Atg18/Atg9 autophagy complex in the maintenance of mitochondrial integrity and, regulation of heart and muscle functions in Drosophila , raising the possibility of augmenting Atg2-Atg18/Atg9 activity in promoting mitochondrial health and, muscle and heart function. Highlights • Knockdown of Atg2, Atg9 or Atg18 in muscle and heart leads to impaired negative geotaxis and a short lifespan in Drosophila. • Knockdown of Atg2, Atg18 or Atg9 causes age-dependent impairment of cardiac structure and function. • Atg2, Atg18 and Atg9 are required in the heart to maintain mitochondrial structure and Drp1 and Mfn2 mRNA expression. • Atg2, Atg18 and Atg9 are required for maintenance of mitochondrial structure and density in IFM. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
144. Bayesian parameter estimation for dynamical models in systems biology
- Author
-
Padmini Rangamani, Boris Kramer, Nathaniel Linden, and Saucerman, Jeffrey J
- Subjects
Ecology ,Bioinformatics ,Molecular Networks (q-bio.MN) ,Systems Biology ,Bayes Theorem ,Bioengineering ,Biological Sciences ,Biological ,Models, Biological ,Quantitative Biology - Quantitative Methods ,Mathematical Sciences ,Kinetics ,Cellular and Molecular Neuroscience ,Computational Theory and Mathematics ,Models ,FOS: Biological sciences ,Information and Computing Sciences ,Modeling and Simulation ,Genetics ,Quantitative Biology - Molecular Networks ,Generic health relevance ,Molecular Biology ,Quantitative Methods (q-bio.QM) ,Ecology, Evolution, Behavior and Systematics ,Signal Transduction - Abstract
Dynamical systems modeling, particularly via systems of ordinary differential equations, has been used to effectively capture the temporal behavior of different biochemical components in signal transduction networks. Despite the recent advances in experimental measurements, including sensor development and '-omics' studies that have helped populate protein-protein interaction networks in great detail, modeling in systems biology lacks systematic methods to estimate kinetic parameters and quantify associated uncertainties. This is because of multiple reasons, including sparse and noisy experimental measurements, lack of detailed molecular mechanisms underlying the reactions, and missing biochemical interactions. Additionally, the inherent nonlinearities with respect to the states and parameters associated with the system of differential equations further compound the challenges of parameter estimation. In this study, we propose a comprehensive framework for Bayesian parameter estimation and complete quantification of the effects of uncertainties in the data and models. We apply these methods to a series of signaling models of increasing mathematical complexity. Systematic analysis of these dynamical systems showed that parameter estimation depends on data sparsity, noise level, and model structure, including the existence of multiple steady states. These results highlight how focused uncertainty quantification can enrich systems biology modeling and enable additional quantitative analyses for parameter estimation., 58 pages, 24 figures
- Published
- 2022
145. Computational modeling of cardiac fibroblasts and fibrosis.
- Author
-
Zeigler, Angela C., Richardson, William J., Holmes, Jeffrey W., and Saucerman, Jeffrey J.
- Subjects
- *
HEART fibrosis , *FIBROBLASTS , *DIASTOLE (Cardiac cycle) , *TARGETED drug delivery , *CELL lines - Abstract
Altered fibroblast behavior can lead to pathologic changes in the heart such as arrhythmia, diastolic dysfunction, and systolic dysfunction. Computational models are increasingly used as a tool to identify potential mechanisms driving a phenotype or potential therapeutic targets against an unwanted phenotype. Here we review how computational models incorporating cardiac fibroblasts have clarified the role for these cells in electrical conduction and tissue remodeling in the heart. Models of fibroblast signaling networks have primarily focused on fibroblast cell lines or fibroblasts from other tissues rather than cardiac fibroblasts, specifically, but they are useful for understanding how fundamental signaling pathways control fibroblast phenotype. In the future, modeling cardiac fibroblast signaling, incorporating -omics and drug-interaction data into signaling network models, and utilizing multi-scale models will improve the ability of in silico studies to predict potential therapeutic targets against adverse cardiac fibroblast activity. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
146. Rate-dependent effects of lidocaine on cardiac dynamics: Development and analysis of a low-dimensional drug-channel interaction model
- Author
-
Colleen E. Clancy, Steffen S. Docken, Timothy J. Lewis, and Saucerman, Jeffrey J
- Subjects
0301 basic medicine ,Patch-Clamp Techniques ,Lidocaine ,Physiology ,Action Potentials ,Voltage-Gated Sodium Channels ,Arrhythmias ,030204 cardiovascular system & hematology ,Cardiovascular ,Quantitative Biology - Quantitative Methods ,Biochemistry ,Mathematical Sciences ,Ion Channels ,Sodium Channels ,0302 clinical medicine ,Sodium channel blocker ,Plateau potentials ,Models ,Heart Rate ,Medicine and Health Sciences ,Drug Interactions ,Biology (General) ,Quantitative Methods (q-bio.QM) ,Membrane potential ,Drug Dependence ,Voltage-Gated Sodium Channel Blockers ,Ecology ,Chemistry ,Approximation Methods ,Physics ,Models, Cardiovascular ,Heart ,Biological Sciences ,Receptor–ligand kinetics ,Markov Chains ,Electrophysiology ,Computational Theory and Mathematics ,5.1 Pharmaceuticals ,Behavioral Pharmacology ,Modeling and Simulation ,Physical Sciences ,Development of treatments and therapeutic interventions ,Cardiac ,Anti-Arrhythmia Agents ,Arrhythmia ,medicine.drug ,Research Article ,Bioinformatics ,QH301-705.5 ,Biophysics ,Cardiology ,Neurophysiology ,Markov model ,Membrane Potential ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Information and Computing Sciences ,Genetics ,medicine ,Animals ,Humans ,Computer Simulation ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,Pharmacology ,Sodium channel ,Myocardium ,Biology and Life Sciences ,Proteins ,Computational Biology ,Interaction model ,Arrhythmias, Cardiac ,Kinetics ,030104 developmental biology ,Algebra ,FOS: Biological sciences ,Algebraic Geometry ,Mathematics ,Neuroscience - Abstract
State-dependent sodium channel blockers are often prescribed to treat cardiac arrhythmias, but many sodium channel blockers are known to have pro-arrhythmic side effects. While the anti and proarrhythmic potential of a sodium channel blocker is thought to depend on the characteristics of its rate-dependent block, the mechanisms linking these two attributes are unclear. Furthermore, how specific properties of rate-dependent block arise from the binding kinetics of a particular drug is poorly understood. Here, we examine the rate-dependent effects of the sodium channel blocker lidocaine by constructing and analyzing a novel drug-channel interaction model. First, we identify the predominant mode of lidocaine binding in a 24 variable Markov model for lidocaine-sodium channel interaction by Moreno et al. Specifically, we find that (1) the vast majority of lidocaine bound to sodium channels is in the neutral form, i.e., the binding of charged lidocaine to sodium channels is negligible, and (2) neutral lidocaine binds almost exclusively to inactivated channels and, upon binding, immobilizes channels in the inactivated state. We then develop a novel 3-variable lidocaine-sodium channel interaction model that incorporates only the predominant mode of drug binding. Our low-dimensional model replicates an extensive amount of the voltage-clamp data used to parameterize the Moreno et al. model. Furthermore, the effects of lidocaine on action potential upstroke velocity and conduction velocity in our model are similar to those predicted by the Moreno et al. model. By exploiting the low-dimensionality of our model, we derive an algebraic expression for level of rate-dependent block as a function of pacing frequency, restitution properties, diastolic and plateau potentials, and drug binding rate constants. Our model predicts that the level of rate-dependent block is sensitive to alterations in restitution properties and increases in diastolic potential, but it is insensitive to variations in the shape of the action potential waveform and lidocaine binding rates., Author summary Cardiac arrhythmias are often treated with drugs that block and alter the kinetics of membrane sodium channels. However, different drugs interact with sodium channels in different ways, and the complexity of the drug-channel interactions makes it difficult to predict whether a particular sodium channel blocker will reduce or increase the probability of cardiac arrhythmias. Here, we characterize the binding kinetics and effects on electrical signal propagation of the antiarrhythmic drug lidocaine, which is an archetypical example of a safe sodium channel blocker. Through analysis of a high-dimensional biophysically-detailed model of lidocaine-sodium channel interaction, we identify the predominant lidocaine binding pathway. We then incorporate only the key features of the predominant binding pathway into a novel low-dimensional model of lidocaine-sodium channel interaction. Our analysis of the low-dimensional model characterizes how the key binding properties of lidocaine affect electrical signal generation and propagation in the heart, and therefore our results are a step towards understanding the features that differentiate pro- and antiarrhythmic sodium channel blockers.
- Published
- 2021
147. Striated myocyte structural integrity: Automated analysis of sarcomeric z-discs
- Author
-
Tessa Altair Morris, Tohru Kiyono, Xiangduo Kong, Kirby Sinclair Fibben, Kyoko Yokomori, Anna Grosberg, Jasmine Naik, and Saucerman, Jeffrey J
- Subjects
0301 basic medicine ,Muscle Physiology ,Physiology ,Computer science ,Image Processing ,Muscle Fibers, Skeletal ,Cardiovascular ,Biochemistry ,Sarcomere ,Mathematical Sciences ,Rats, Sprague-Dawley ,Contractile Proteins ,0302 clinical medicine ,Computer-Assisted ,Myofibrils ,Animal Cells ,Medicine and Health Sciences ,Image Processing, Computer-Assisted ,Myocyte ,Myocytes, Cardiac ,Biology (General) ,Cells, Cultured ,Cardiomyocytes ,Microscopy ,Cultured ,Ecology ,Quality assessment ,Physics ,Skeletal ,Biological Sciences ,Condensed Matter Physics ,Aspect Ratio ,Heart Disease ,Computational Theory and Mathematics ,Modeling and Simulation ,Physical Sciences ,Muscle ,Cellular Types ,Anatomy ,Cardiac ,Algorithms ,Research Article ,Muscle Contraction ,Sarcomeres ,QH301-705.5 ,Bioinformatics ,Cells ,Materials Science ,Material Properties ,Muscle Tissue ,Geometry ,Bioengineering ,Striated Muscles ,Muscle Fibers ,Fluorescence ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Information and Computing Sciences ,Genetics ,Animals ,Humans ,Muscle, Skeletal ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,Actin ,Muscle Cells ,Myocytes ,Biology and Life Sciences ,Proteins ,Structural integrity ,Cell Biology ,Actins ,Current analysis ,Rats ,Cytoskeletal Proteins ,Biological Tissue ,030104 developmental biology ,Microscopy, Fluorescence ,Anisotropy ,Sprague-Dawley ,Myofibril ,Neuroscience ,Mathematics ,030217 neurology & neurosurgery - Abstract
As sarcomeres produce the force necessary for contraction, assessment of sarcomere order is paramount in evaluation of cardiac and skeletal myocytes. The uniaxial force produced by sarcomeres is ideally perpendicular to their z-lines, which couple parallel myofibrils and give cardiac and skeletal myocytes their distinct striated appearance. Accordingly, sarcomere structure is often evaluated by staining for z-line proteins such as α-actinin. However, due to limitations of current analysis methods, which require manual or semi-manual handling of images, the mechanism by which sarcomere and by extension z-line architecture can impact contraction and which characteristics of z-line architecture should be used to assess striated myocytes has not been fully explored. Challenges such as isolating z-lines from regions of off-target staining that occur along immature stress fibers and cell boundaries and choosing metrics to summarize overall z-line architecture have gone largely unaddressed in previous work. While an expert can qualitatively appraise tissues, these challenges leave researchers without robust, repeatable tools to assess z-line architecture across different labs and experiments. Additionally, the criteria used by experts to evaluate sarcomeric architecture have not been well-defined. We address these challenges by providing metrics that summarize different aspects of z-line architecture that correspond to expert tissue quality assessment and demonstrate their efficacy through an examination of engineered tissues and single cells. In doing so, we have elucidated a mechanism by which highly elongated cardiomyocytes become inefficient at producing force. Unlike previous manual or semi-manual methods, characterization of z-line architecture using the metrics discussed and implemented in this work can quantitatively evaluate engineered tissues and contribute to a robust understanding of the development and mechanics of striated muscles., Author summary Structural evaluation of sarcomeres is fundamental to the study of striated muscle. However, due to limitations of current analysis methods, the mechanisms by which sarcomere order can impact contraction and the characteristics of sarcomere architecture that should be used to assess striated myocytes have not been fully explored. Furthermore, it is not clear what aspects of sarcomere architecture are considered by the experts when qualitatively evaluating striated muscle tissues. Therefore, we developed a computational structural assay in MATLAB, ZlineDetection, to evaluate sarcomere architecture by both extracting sarcomeric z-lines from images and providing metrics that encapsulate different aspects of z-line architecture that an expert would evaluate when judging the quality of the tissue. The sarcomere structure of both patient-specific skeletal muscle and rat cardiomyocytes were evaluated with differences among engineered cells and tissues quantified using novel and established metrics. As a result, a mechanism by which highly elongated cardiomyocytes become inefficient at producing force was elucidated. ZlineDetection identifies and quantifies the characteristics used by experts for evaluation and thus it will lead to more rigorous differentiation methods and tissue comparison across labs and contribute a robust understanding of how structure affects mechanical function.
- Published
- 2020
148. Nanoscale organization of ryanodine receptor distribution and phosphorylation pattern determines the dynamics of calcium sparks
- Author
-
María Hernández Mesa, Jonas van den Brink, William E. Louch, Kimberly J. McCabe, Padmini Rangamani, and Saucerman, Jeffrey J
- Subjects
Heart Failure ,Myocytes ,Ecology ,Bioinformatics ,Biophysics ,Ryanodine Receptor Calcium Release Channel ,Biological Sciences ,Cardiovascular ,Mathematical Sciences ,Cellular and Molecular Neuroscience ,Sarcoplasmic Reticulum ,Heart Disease ,Computational Theory and Mathematics ,Modeling and Simulation ,Information and Computing Sciences ,Genetics ,Humans ,Myocytes, Cardiac ,Calcium ,Calcium Signaling ,Phosphorylation ,Molecular Biology ,Cardiac ,Ecology, Evolution, Behavior and Systematics - Abstract
Super-resolution imaging techniques have provided a better understanding of the relationship between the nanoscale organization and function of ryanodine receptors (RyRs) in cardiomyocytes. Recent data have indicated that this relationship is disrupted in heart failure (HF), as RyRs are dispersed into smaller and more numerous clusters. However, RyRs are also hyperphosphorylated in this condition, and this is reported to occur preferentially within the cluster centre. Thus, the combined impact of RyR relocalization and sensitization on Ca2+ spark generation in failing cardiomyocytes is likely complex and these observations suggest that both the nanoscale organization of RyRs and the pattern of phosphorylated RyRs within clusters could be critical determinants of Ca2+ spark dynamics. To test this hypothesis, we used computational modeling to quantify the relationships between RyR cluster geometry, phosphorylation patterns, and sarcoplasmic reticulum (SR) Ca2+ release. We found that RyR cluster disruption results in a decrease in spark fidelity and longer sparks with a lower amplitude. Phosphorylation of some RyRs within the cluster can play a compensatory role, recovering healthy spark dynamics. Interestingly, our model predicts that such compensation is critically dependent on the phosphorylation pattern, as phosphorylation localized within the cluster center resulted in longer Ca2+ sparks and higher spark fidelity compared to a uniformly distributed phosphorylation pattern. Our results strongly suggest that both the phosphorylation pattern and nanoscale RyR reorganization are critical determinants of Ca2+ dynamics in HF.
- Published
- 2022
149. Computational modeling of blood component transport related to coronary artery thrombosis in Kawasaki disease
- Author
-
Noelia Grande Gutierrez, Brian W. McCrindle, Alison L. Marsden, Mark Alber, Jane C. Burns, Andrew M. Kahn, Mathew Mathew, and Saucerman, Jeffrey J
- Subjects
Platelet Aggregation ,Physiology ,Epidemiology ,Hemodynamics ,Cardiovascular Medicine ,Cardiovascular ,Vascular Medicine ,Mathematical Sciences ,chemistry.chemical_compound ,Medical Conditions ,Animal Cells ,Medicine and Health Sciences ,Medicine ,Platelet ,Biology (General) ,Coronary Arteries ,Ecology ,Arteries ,Hematology ,Biological Sciences ,Coronary Vessels ,Thrombosis ,Body Fluids ,Adenosine Diphosphate ,Heart Disease ,Blood ,medicine.anatomical_structure ,Computational Theory and Mathematics ,Coagulation ,Cardiovascular Diseases ,Modeling and Simulation ,Cardiology ,Anatomy ,Cellular Types ,Aneurysms ,Research Article ,Artery ,Platelets ,Blood Platelets ,medicine.medical_specialty ,QH301-705.5 ,Bioinformatics ,Mucocutaneous Lymph Node Syndrome ,Cellular and Molecular Neuroscience ,Clinical Research ,Information and Computing Sciences ,Internal medicine ,Genetics ,Humans ,Computer Simulation ,Vascular Diseases ,Platelet activation ,Blood Coagulation ,Molecular Biology ,Heart Disease - Coronary Heart Disease ,Ecology, Evolution, Behavior and Systematics ,Blood Cells ,Coagulation Disorders ,business.industry ,Biology and Life Sciences ,Anticoagulants ,Computational Biology ,Cell Biology ,Platelet Activation ,medicine.disease ,Adenosine diphosphate ,chemistry ,Medical Risk Factors ,Cardiovascular Anatomy ,Blood Vessels ,Kawasaki disease ,business - Abstract
Coronary artery thrombosis is the major risk associated with Kawasaki disease (KD). Long-term management of KD patients with persistent aneurysms requires a thrombotic risk assessment and clinical decisions regarding the administration of anticoagulation therapy. Computational fluid dynamics has demonstrated that abnormal KD coronary artery hemodynamics can be associated with thrombosis. However, the underlying mechanisms of clot formation are not yet fully understood. Here we present a new model incorporating data from patient-specific simulated velocity fields to track platelet activation and accumulation. We use a system of Reaction-Advection-Diffusion equations solved with a stabilized finite element method to describe the evolution of non-activated platelets and activated platelet concentrations [AP], local concentrations of adenosine diphosphate (ADP) and poly-phosphate (PolyP). The activation of platelets is modeled as a function of shear-rate exposure and local concentration of agonists. We compared the distribution of activated platelets in a healthy coronary case and six cases with coronary artery aneurysms caused by KD, including three with confirmed thrombosis. Results show spatial correlation between regions of higher concentration of activated platelets and the reported location of the clot, suggesting predictive capabilities of this model towards identifying regions at high risk for thrombosis. Also, the concentration levels of ADP and PolyP in cases with confirmed thrombosis are higher than the reported critical values associated with platelet aggregation (ADP) and activation of the intrinsic coagulation pathway (PolyP). These findings suggest the potential initiation of a coagulation pathway even in the absence of an extrinsic factor. Finally, computational simulations show that in regions of flow stagnation, biochemical activation, as a result of local agonist concentration, is dominant. Identifying the leading factors to a pro-coagulant environment in each case—mechanical or biochemical—could help define improved strategies for thrombosis prevention tailored for each patient., Author Summary Computational studies aiming to model thrombosis often rely on an arterial wall injury. Collagen and other extracellular matrix components are exposed to the bloodstream, which facilitates platelet adhesion to the wall and subsequent clot formation. However, these models are not adequate to explain thrombosis in other settings where even in the absence of a focal lesion, clots may still form under certain flow conditions. Coronary artery aneurysm thrombosis following KD is an example of the need to understand the mechanisms of thrombus initiation in the absence of an extrinsic factor. This study provides a new framework to investigate thrombus initiation in KD from a patient-specific perspective, which integrates fluid mechanics and biochemistry and which could help quantify the pro-coagulant environment induced by the aneurysm and become a predictive tool. The work presented here has broad relevance to other clinical situations where flow stagnation and transport are driving factors in thrombus formation.
- Published
- 2021
150. Long-Lasting Sparks: Multi-Metastability and Release Competition in the Calcium Release Unit Network
- Author
-
James N. Weiss, Alain Karma, Zhen Song, Zhilin Qu, and Saucerman, Jeffrey J
- Subjects
0301 basic medicine ,Long lasting ,Bioinformatics ,chemistry.chemical_element ,Calcium ,Models, Biological ,Mathematical Sciences ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Models ,Information and Computing Sciences ,Metastability ,Genetics ,Calcium Signaling ,lcsh:QH301-705.5 ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,Noise strength ,Simulation ,Calcium signaling ,Physics ,Ecology ,Voltage-dependent calcium channel ,Ryanodine receptor ,Computational Biology ,Ryanodine Receptor Calcium Release Channel ,Biological Sciences ,Biological ,Sarcoplasmic Reticulum ,030104 developmental biology ,Critical level ,lcsh:Biology (General) ,Computational Theory and Mathematics ,chemistry ,Modeling and Simulation ,Biophysics ,Research Article - Abstract
Calcium (Ca) sparks are elementary events of biological Ca signaling. A normal Ca spark has a brief duration in the range of 10 to 100 ms, but long-lasting sparks with durations of several hundred milliseconds to seconds are also widely observed. Experiments have shown that the transition from normal to long-lasting sparks can occur when ryanodine receptor (RyR) open probability is either increased or decreased. Here, we demonstrate theoretically and computationally that long-lasting sparks emerge as a collective dynamical behavior of the network of diffusively coupled Ca release units (CRUs). We show that normal sparks occur when the CRU network is monostable and excitable, while long-lasting sparks occur when the network dynamics possesses multiple metastable attractors, each attractor corresponding to a different spatial firing pattern of sparks. We further highlight the mechanisms and conditions that produce long-lasting sparks, demonstrating the existence of an optimal range of RyR open probability favoring long-lasting sparks. We find that when CRU firings are sparse and sarcoplasmic reticulum (SR) Ca load is high, increasing RyR open probability promotes long-lasting sparks by potentiating Ca-induced Ca release (CICR). In contrast, when CICR is already strong enough to produce frequent firings, decreasing RyR open probability counter-intuitively promotes long-lasting sparks by decreasing spark frequency. The decrease in spark frequency promotes intra-SR Ca diffusion from neighboring non-firing CRUs to the firing CRUs, which helps to maintain the local SR Ca concentration of the firing CRUs above a critical level to sustain firing. In this setting, decreasing RyR open probability further suppresses long-lasting sparks by weakening CICR. Since a long-lasting spark terminates via the Kramers’ escape process over a potential barrier, its duration exhibits an exponential distribution determined by the barrier height and noise strength, which is modulated differently by different ways of altering the Ca release flux strength., Author Summary Calcium (Ca) sparks, resulting from Ca-induced Ca release, are elementary events of biological Ca signaling. Sparks are normally brief, but long-lasting sparks have been widely observed experimentally under various conditions. The underlying mechanisms of spark duration or termination and the corresponding determinants remain a topic of debate. In this study, we demonstrate theoretically and computationally that normal brief sparks are excitable transients, while long-lasting sparks are multiple metastable states emerging in the diffusively coupled Ca release unit network, as a result of cooperativity and release competition among the Ca release units. Termination of a long-lasting spark is a Kramers’ escape process over a potential barrier, and the spark duration is the first-passage time, exhibiting an exponential distribution.
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.